1
|
Geisert RD, Bazer FW, Lucas CG, Pfeiffer CA, Meyer AE, Sullivan R, Johns DN, Sponchiado M, Prather RS. Maternal recognition of pregnancy in the pig: A servomechanism involving sex steroids, cytokines and prostaglandins. Anim Reprod Sci 2024; 264:107452. [PMID: 38522133 DOI: 10.1016/j.anireprosci.2024.107452] [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: 01/30/2024] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/26/2024]
Abstract
Maternal recognition of pregnancy (MRP) is a term utilized in mammals to describe pathways in which the conceptus alters the endometrial environment to prevent regression of corpora lutea to ensure continued production of progesterone (P4) required for establishment and maintenance of pregnancy. For nearly 40 years after publication of the endocrine/exocrine theory, conceptus estrogen (E2) was considered the primary maternal recognition signal in the pig. Conceptus production of prostaglandin E2 (PGE2) was also considered to be a major factor in preventing luteolysis. An addition to E2 and PGE2, pig conceptuses produce interleukin 1B2 (IL1B2) and interferons (IFN) delta (IFND) and gamma (IFNG). The present review provides brief history of the discovery of E2, PGs and IFNS which led to research investigating the role of these conceptus secreted factors in establishing and maintaining pregnancy in the pig. The recent utilization of gene editing technology allowed a more direct approach to investigate the in vivo roles of IL1B2, E2, PGE2, AND IFNG for establishment of pregnancy. These studies revealed unknown functions for IFNG and ILB2 in addition to PGE2 and E2. Thus, pregnancy recognition signal is via a servomechanism in requiring sequential effects of P4, E2, IL1B2, PGE2 and IFNG. Results indicate that the original established dogma for the role of conceptus E2 and PGs in MRP is a far too simplified model that involves the interplay of numerous mechanisms for inhibiting luteolysis, inducing critical elongation of the conceptuses and resolution of inflammation in pigs.
Collapse
Affiliation(s)
- Rodney D Geisert
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA.
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA
| | - Caroline G Lucas
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Caroline A Pfeiffer
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Ashley E Meyer
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Riley Sullivan
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Destiny N Johns
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Mariana Sponchiado
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Randall S Prather
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
2
|
Aleem AM, Kang W, Lin S, Milad M, Kingsley PJ, Crews BC, Uddin MJ, Rouzer CA, Marnett LJ. Ferroptosis Inhibitors Suppress Prostaglandin Synthesis in Lipopolysaccharide-Stimulated Macrophages. ACS Chem Biol 2023; 18:404-418. [PMID: 36638351 DOI: 10.1021/acschembio.2c00869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Necrostatin-1 blocks ferroptosis via an unknown mechanism and necroptosis through inhibition of receptor-interacting protein kinase-1 (RIP1). We report that necrostatin-1 suppresses cyclooxygenase-2-dependent prostaglandin biosynthesis in lipopolysaccharide-treated RAW264.7 macrophages (IC50 ∼ 100 μM). This activity is shared by necrostatin-1i (IC50 ∼ 50 μM), which lacks RIP1 inhibitory activity, but not the RIP1 inhibitors necrostatin-1s or deschloronecrostatin-1s. Furthermore, we show that the potent ferroptosis inhibitors and related compounds ferrostatin-1, phenoxazine, phenothiazine, and 10-methylphenothiazine strongly inhibit cellular prostaglandin biosynthesis with IC50's in the range of 30 nM to 3.5 μM. None of the compounds inhibit lipopolysaccharide-mediated cyclooxygenase-2 protein induction. In the presence of activating hydroperoxides, the necrostatins and ferroptosis inhibitors range from low potency inhibition to stimulation of in vitro cyclooxygenase-2 activity; however, inhibitory potency is increased under conditions of low peroxide tone. The ferroptosis inhibitors are highly effective reducing substrates for cyclooxygenase-2's peroxidase activity, suggesting that they act by suppressing hydroperoxide-mediated activation of the cyclooxygenase active site. In contrast, for the necrostatins, cellular prostaglandin synthesis inhibition does not correlate with peroxidase-reducing activity but rather with the presence of a thiohydantoin substituent, which conveys the ability to reduce the endoperoxide intermediate prostaglandin H2 to prostaglandin F2α in vitro. This finding suggests that necrostatin-1 blocks cellular prostaglandin synthesis and ferroptosis via a redox mechanism distinct from action as a one-electron donor. The results indicate that a wide range of compounds derived from redox-active chemical scaffolds can block cellular prostaglandin biosynthesis.
Collapse
Affiliation(s)
- Ansari M Aleem
- A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Weixi Kang
- A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Shuyang Lin
- A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Matthew Milad
- A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Philip J Kingsley
- A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Brenda C Crews
- A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Md Jashim Uddin
- A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Carol A Rouzer
- A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, and 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, and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
3
|
Alvarez MS, Núñez E, Fuertes-Agudo M, Cucarella C, Fernandez-Velasco M, Boscá L, Vázquez J, Rossignol R, Martin-Sanz P, Casado M. Quantitative Proteomics Analysis Reveals That Cyclooxygenase-2 Modulates Mitochondrial Respiratory Chain Complex IV in Cardiomyocytes. Int J Mol Sci 2022; 23:13476. [PMID: 36362254 PMCID: PMC9655412 DOI: 10.3390/ijms232113476] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/24/2022] [Accepted: 10/30/2022] [Indexed: 10/10/2023] Open
Abstract
The biochemical mechanisms of cell injury and myocardial cell death after myocardial infarction remain unresolved. Cyclooxygenase 2 (COX-2), a key enzyme in prostanoid synthesis, is expressed in human ischemic myocardium and dilated cardiomyopathy, but it is absent in healthy hearts. To assess the role of COX-2 in cardiovascular physiopathology, we developed transgenic mice that constitutively express functional human COX-2 in cardiomyocytes under the control of the α-myosin heavy chain promoter. These animals had no apparent phenotype but were protected against ischemia-reperfusion injury in isolated hearts, with enhanced functional recovery and diminished cellular necrosis. To further explore the phenotype of this animal model, we carried out a differential proteome analysis of wild-type vs. transgenic cardiomyocytes. The results revealed a tissue-specific proteomic profile dominated by mitochondrial proteins. In particular, an increased expression of respiratory chain complex IV proteins was observed. This correlated with increased catalytic activity, enhanced respiratory capacity, and increased ATP levels in the heart of COX-2 transgenic mice. These data suggest a new link between COX-2 and mitochondria, which might contribute to the protective cardiac effects of COX-2 against ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Maria Soledad Alvarez
- Instituto de Biomedicina de Valencia (IBV), CSIC, Jaume Roig 11, 46010 Valencia, Spain
| | - Estefanía Núñez
- Laboratory of Cardiovascular Proteomics, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Marina Fuertes-Agudo
- Instituto de Biomedicina de Valencia (IBV), CSIC, Jaume Roig 11, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Carme Cucarella
- Instituto de Biomedicina de Valencia (IBV), CSIC, Jaume Roig 11, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Maria Fernandez-Velasco
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Monforte de Lemos 3-5, 28029 Madrid, Spain
- Instituto de Investigación Hospital Universitario La Paz, IDIPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
| | - Lisardo Boscá
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Monforte de Lemos 3-5, 28029 Madrid, Spain
- Instituto de Investigaciones Biomedicas Alberto Sols (IIBM), CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain
| | - Jesús Vázquez
- Laboratory of Cardiovascular Proteomics, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Rodrigue Rossignol
- Laboratoire Maladies Rares, CHU Pellegrin Place Amelie Rab, 33076 Bordeaux, France
| | - Paloma Martin-Sanz
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
- Instituto de Investigaciones Biomedicas Alberto Sols (IIBM), CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain
| | - Marta Casado
- Instituto de Biomedicina de Valencia (IBV), CSIC, Jaume Roig 11, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
| |
Collapse
|
4
|
Modulation of Enzyme-Catalyzed Synthesis of Prostaglandins by Components Contained in Kidney Microsomal Preparations. MOLECULES (BASEL, SWITZERLAND) 2021; 27:molecules27010219. [PMID: 35011450 PMCID: PMC8746486 DOI: 10.3390/molecules27010219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 11/17/2022]
Abstract
In the kidney, prostaglandins formed by cyclooxygenase 1 and 2 (COX-1 and COX-2) play an important role in regulating renal blood flow. In the present study, we report our observations regarding a unique modulatory effect of renal microsomal preparation on COX-1/2-mediated formation of major prostaglandin (PG) products in vitro. We found that microsomes prepared from pig and rat kidneys had a dual stimulatory–inhibitory effect on the formation of certain PG products catalyzed by COX-1 and COX-2. At lower concentrations, kidney microsomes stimulated the formation of certain PG products, whereas at higher concentrations, their presence inhibited the formation. Presence of kidney microsomes consistently increased the Km values of the COX-1/2-mediated reactions, while the Vmax might be increased or decreased depending on stimulation or inhibition observed. Experimental evidence was presented to show that a protein component present in the pig kidney microsomes was primarily responsible for the activation of the enzyme-catalyzed arachidonic acid metabolism leading to the formation of certain PG products.
Collapse
|
5
|
Figurová D, Tokárová K, Greifová H, Knížatová N, Kolesárová A, Lukáč N. Inflammation, It's Regulation and Antiphlogistic Effect of the Cyanogenic Glycoside Amygdalin. Molecules 2021; 26:5972. [PMID: 34641516 PMCID: PMC8512454 DOI: 10.3390/molecules26195972] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 12/14/2022] Open
Abstract
The inflammatory reaction accompanies in part or in full any disease process in the vascularized metazoan. This complicated reaction is controlled by regulatory mechanisms, some of which produce unpleasant symptomatic manifestations of inflammation. Therefore, there has been an effort to develop selective drugs aimed at removing pain, fever, or swelling. Gradually, however, serious adverse side effects of such inhibitors became apparent. Scientific research has therefore continued to explore new possibilities, including naturally available substances. Amygdalin is a cyanogenic glycoside present, e.g., in bitter almonds. This glycoside has already sparked many discussions among scientists, especially about its anticancer potential and related toxic cyanides. However, toxicity at different doses made it generally unacceptable. Although amygdalin given at the correct oral dose may not lead to poisoning, it has not yet been accurately quantified, as its action is often affected by different intestinal microbial consortia. Its pharmacological activities have been studied, but its effects on the body's inflammatory response are lacking. This review discusses the chemical structure, toxicity, and current knowledge of the molecular mechanism of amygdalin activity on immune functions, including the anti-inflammatory effect, but also discusses inflammation as such, its mediators with diverse functions, which are usually targeted by drugs.
Collapse
Affiliation(s)
| | - Katarína Tokárová
- Department of Animal Physiology, Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Trieda Andreja Hlinku 2, 949 76 Nitra, Slovakia; (D.F.); (H.G.); (N.K.); (A.K.); (N.L.)
| | | | | | | | | |
Collapse
|
6
|
Yang C, Li P, Wang P, Zhu BT. Mechanism of reactivation of the peroxidase catalytic activity of human cyclooxygenases by reducing cosubstrate quercetin. J Mol Graph Model 2021; 107:107941. [PMID: 34091174 DOI: 10.1016/j.jmgm.2021.107941] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 04/22/2021] [Accepted: 05/11/2021] [Indexed: 11/19/2022]
Abstract
Our earlier studies show that the peroxidase activity of cyclooxygenase 1 and 2 (COX-1 and COX-2) can be reactivated in vitro and in vivo by the presence of certain naturally-occurring flavonoids such as quercetin and myricetin, which serve as reducing cosubstrates. These compounds can activate COX at nanomolar concentrations. In the present study, quercetin is used as a representative model compound to investigate the chemical mechanism by which the peroxidase activity of human COX-1 and COX-2 is reactivated after each catalytic cycle. Molecular docking and quantum mechanics calculations are carried out to probe the interactions of quercetin with the peroxidase sites of COX-1/2 and the reactivation mechanism. It is found that some of the partially-ionized states of quercetin can bind tightly and closely inside the peroxidase active sites of the COX enzymes and directly interact with heme Fe ion. While quercetin contains several phenolic hydroxyl groups, it is found that only the C-3'-OH group can effectively donate an electron for the reduction of heme because it not only can bind closely and tightly inside the peroxidase sites of COX-1/2, but it can also facilely donate an electron to heme Fe ion. This investigation provides a mechanistic explanation for the chemical process by which quercetin reactivates COX-1/2 peroxidases. This knowledge would aid in the rational design of drugs that can selectively target the peroxidase sites of COX-1/2 either as activators or inhibitors.
Collapse
Affiliation(s)
- Chengxi Yang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Peng Li
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Pan Wang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China; Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China; Shenzhen Bay Laboratory, Shenzhen 518055, China.
| |
Collapse
|
7
|
Bai HW, Yang C, Wang P, Rao S, Zhu BT. Inhibition of cyclooxygenase by blocking the reducing cosubstrate at the peroxidase site: Discovery of galangin as a novel cyclooxygenase inhibitor. Eur J Pharmacol 2021; 899:174036. [PMID: 33737009 DOI: 10.1016/j.ejphar.2021.174036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/01/2022]
Abstract
Earlier we have shown that certain flavonoids (e.g., quercetin) are high-affinity reducing cosubstrates for cyclooxygenase (COX) 1 and 2. These compounds can bind inside the peroxidase active sites of COXs and donate an electron from one of their B-ring hydroxyl groups to hematin. Based on these earlier findings, it is postulated that some of the natural flavonoids such as galangin that are structural analogs of quercetin but lack the proper B-ring hydroxyl groups might function as novel inhibitors of COXs by blocking the effect of the reducing cosubstrates. This idea is tested in the present study. Computational docking analysis together with quantum chemistry calculation shows that galangin can bind inside the peroxidase active sites of COX-1 and COX-2 in a similar manner as quercetin, but it has little ability to effectively donate its electrons, thereby blocking the effect of the reducing cosubstrates like quercetin. Further experimental studies confirm that galangin can inhibit, both in vitro and in vivo, quercetin-mediated activation of the peroxidase activity of the COX-1/2 enzymes. The results of the present study demonstrate that galangin is a novel naturally-occurring inhibitor of COX-1 and COX-2, acting by blocking the function of the reducing cosubstrates at the peroxidase sites.
Collapse
Affiliation(s)
- Hyoung-Woo Bai
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China; Present Address: Advanced Radiation Technology Institute (ARTI), Korea Atomic Energy Research Institute (KAERI), Jeongeup-si, Jeollabuk-do 580-185, Republic of Korea
| | - Chengxi Yang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China
| | - Pan Wang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China
| | - Shun Rao
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China; Department of Pharmacology, Toxicology and Therapeutics, School of Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
8
|
Murru E, Carta G, Manca C, Sogos V, Pistis M, Melis M, Banni S. Conjugated Linoleic Acid and Brain Metabolism: A Possible Anti-Neuroinflammatory Role Mediated by PPARα Activation. Front Pharmacol 2021; 11:587140. [PMID: 33505308 PMCID: PMC7832089 DOI: 10.3389/fphar.2020.587140] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/17/2020] [Indexed: 12/24/2022] Open
Abstract
Fatty acids play a crucial role in the brain as specific receptor ligands and as precursors of bioactive metabolites. Conjugated linoleic acid (CLA), a group of positional and geometric isomers of linoleic acid (LA, 18:2 n-6) present in meat and dairy products of ruminants and synthesized endogenously in non-ruminants and humans, has been shown to possess different nutritional properties associated with health benefits. Its ability to bind to peroxisome proliferator-activated receptor (PPAR) α, a nuclear receptor key regulator of fatty acid metabolism and inflammatory responses, partly mediates these beneficial effects. CLA is incorporated and metabolized into brain tissue where induces the biosynthesis of endogenous PPARα ligands palmitoylethanolamide (PEA) and oleoylethanolamide (OEA), likely through a positive feedback mechanism where PPARα activation sustains its own cellular effects through ligand biosynthesis. In addition to PPARα, PEA and OEA may as well bind to other receptors such as TRPV1, further extending CLA own anti-neuroinflammatory actions. Future studies are needed to investigate whether dietary CLA may exert anti-inflammatory activity, particularly in the setting of neurodegenerative diseases and neuropsychiatric disorders with a neuroinflammatory basis.
Collapse
Affiliation(s)
- Elisabetta Murru
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Gianfranca Carta
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Claudia Manca
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Valeria Sogos
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Marco Pistis
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy.,Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| | - Miriam Melis
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Sebastiano Banni
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| |
Collapse
|
9
|
Yamamoto K, Ueda N. In Memoriam: Shozo Yamamoto (1933–2020). J Lipid Res 2020. [DOI: 10.1194/jlr.t120001104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
10
|
Yang C, Li P, Ding X, Sui HC, Rao S, Hsu CH, Leung WP, Cheng GJ, Wang P, Zhu BT. Mechanism for the reactivation of the peroxidase activity of human cyclooxygenases: investigation using phenol as a reducing cosubstrate. Sci Rep 2020; 10:15187. [PMID: 32938962 PMCID: PMC7494923 DOI: 10.1038/s41598-020-71237-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/10/2020] [Indexed: 01/05/2023] Open
Abstract
It has been known for many years that the peroxidase activity of cyclooxygenase 1 and 2 (COX-1 and COX-2) can be reactivated in vitro by the presence of phenol, which serves as a reducing compound, but the underlying mechanism is still poorly understood. In the present study, we use phenol as a model compound to investigate the mechanism by which the peroxidase activity of human COXs is reactivated after each catalytic cycle. Molecular docking and quantum mechanics calculations are carried out to probe the interaction of phenol with the peroxidase site of COXs and the reactivation mechanism. It is found that the oxygen atom associated with the Fe ion in the heme group (i.e., the complex of Fe ion and porphyrin) of COXs can be removed by addition of two protons. Following its removal, phenol can readily bind inside the peroxidase active sites of the COX enzymes, and directly interact with Fe in heme to facilitate electron transfer from phenol to heme. This investigation provides theoretical evidence for several intermediates formed in the COX peroxidase reactivation cycle, thereby unveiling mechanistic details that would aid in future rational design of drugs that target the peroxidase site.
Collapse
Affiliation(s)
- Chengxi Yang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, The Chinese University of Hong Kong, Shenzhen, 518172, China.,School of Life and Health Sciences, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Peng Li
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, The Chinese University of Hong Kong, Shenzhen, 518172, China.,School of Life and Health Sciences, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Xiaoli Ding
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Hao Chen Sui
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Shun Rao
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Chia-Hsiang Hsu
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Wing-Por Leung
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, The Chinese University of Hong Kong, Shenzhen, 518172, China.,School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Gui-Juan Cheng
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, The Chinese University of Hong Kong, Shenzhen, 518172, China.,School of Life and Health Sciences, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Pan Wang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, The Chinese University of Hong Kong, Shenzhen, 518172, China. .,School of Life and Health Sciences, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China. .,Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, The Chinese University of Hong Kong, Shenzhen, 518172, China. .,School of Life and Health Sciences, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China. .,Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| |
Collapse
|
11
|
Kelley KW, Peng YP, Liu Q, Chang HC, Spencer SJ, Hutchinson MR, Shimada A. Psychoneuroimmunology goes East: Development of the PNIRS China affiliate and its expansion into PNIRS Asia-Pacific. Brain Behav Immun 2020; 88:75-87. [PMID: 32304882 PMCID: PMC7156953 DOI: 10.1016/j.bbi.2020.04.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 04/09/2020] [Indexed: 12/14/2022] Open
Abstract
The Psychoneuroimmunology Research Society (PNIRS) created an official Chinese regional affiliate in 2012, designated PNIRSChina. Now, just eight years later, the program has been so successful in advancing the science of psychoneuroimmunology that it has expanded to the whole of Asia-Oceania. In 2017, PNIRSChina became PNIRSAsia-Pacific. Between 2012 and 2019, this outreach affiliate of PNIRS organized seven symposia at major scientific meetings in China as well as nine others in Taiwan, Japan, South Korea, Australia and New Zealand. This paper summarizes the remarkable growth of PNIRSAsia-Pacific. Here, regional experts who have been instrumental in organizing these PNIRSAsia-Pacific symposia briefly review and share their views about the past, present and future state of psychoneuroimmunology research in China, Taiwan, Australia and Japan. The newest initiative of PNIRSAsia-Pacific is connecting Asia-Pacific laboratories with those in Western countries through a simple web-based registration system. These efforts not only contribute to the efforts of PNIRS to serve a truly global scientific society but also to answer the imperative call of increasing diversity in our science.
Collapse
Affiliation(s)
- Keith W Kelley
- Department of Pathology, College of Medicine and Department of Animal Sciences, College of ACES, University of Illinois at Urbana-Champaign, 212 Edward R. Madigan Laboratory, 1201 West Gregory Drive, Urbana, IL 61801, USA.
| | - Yu-Ping Peng
- Department of Physiology and Laboratory of Neuroimmunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, People's Republic of China
| | - Quentin Liu
- Dalian Medical University, Institute of Cancer Stem Cell, Cancer Center Room 317, 9 Lvshun Road South, Dalian 116000, People's Republic of China
| | - Hui-Chih Chang
- Mind-Body Interface Center, China Medical University Hospital, Taichung, Taiwan
| | - Sarah J Spencer
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Mark R Hutchinson
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5005, Australia and the ARC Centre of Excellence for Nanoscale Biophotonics
| | - Atsuyoshi Shimada
- Faculty of Health Sciences, Kyorin University, 5-4-1 Shimorenjaku, Mitaka, 181-8612 Tokyo, Japan
| |
Collapse
|
12
|
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: 86] [Impact Index Per Article: 17.2] [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
|
13
|
Biringer RG. The enzymology of the human prostanoid pathway. Mol Biol Rep 2020; 47:4569-4586. [PMID: 32430846 DOI: 10.1007/s11033-020-05526-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/14/2020] [Indexed: 12/24/2022]
Abstract
Prostanoids are short-lived autocrine and paracrine signaling molecules involved in a wide range of biological functions. They have been shown to be intimately involved in many different disease states when their regulation becomes dysfunctional. In order to fully understand the progression of any disease state or the biological functions of the well state, a complete evaluation of the genomics, proteomics, and metabolomics of the system is necessary. This review is focused on the enzymology for the enzymes involved in the synthesis of the prostanoids (prostaglandins, prostacyclins and thromboxanes). In particular, the isolation and purification of the enzymes, their enzymatic parameters and catalytic mechanisms are presented.
Collapse
Affiliation(s)
- Roger Gregory Biringer
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA.
| |
Collapse
|
14
|
Abstract
Omega-6 polyunsaturated fatty acids were identified as essential nutrients in 1930. Their essentiality is largely due to their function as prostaglandin (PG) precursors. I spent most of my career in biochemistry determining how PG biosynthesis is regulated. PGs are lipid mediators formed in response to certain circulating hormones and cytokines. PGs act near their sites of synthesis to signal neighboring cells to coordinate their responses (e.g. when platelets interact with blood vessels). The committed step in PG synthesis is the conversion of a 20-carbon omega-6 fatty acid called arachidonic acid to prostaglandin endoperoxide H2 (PGH2). Depending on the tissue and the hormone or cytokine stimulus, this reaction is catalyzed by either cyclooxygenase-1 or cyclooxygenase-2 (COX-1 or COX-2). Once formed, PGH2 is converted, again depending on the context, to one of several downstream PG subtypes that act via specific G protein-coupled receptors. Nonsteroidal anti-inflammatory drugs (e.g. aspirin, ibuprofen, and naproxen) block PG synthesis by inhibiting COX-1 and COX-2. COX-2 is also inhibited by COX-2-selective inhibitors. Inhibition of COX-1 by low-dose aspirin prevents thrombosis. COX-2 inhibition reduces inflammation and pain. Investigating the mysteries of COXs anchored my scientific career. I attribute my successes to the great good fortune of having been surrounded by people who helped me make the most of my talents. I have written this reflection in a light-hearted fashion as a self-help essay, while highlighting the people and factors that most impacted me during my upbringing and then during my maturation and evolution as a biochemist.
Collapse
Affiliation(s)
- William L Smith
- From the Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109-0606
| |
Collapse
|
15
|
Dunford HB. Kinetics and Mechanisms of Mammalian Heme Peroxidase Reactions. PROGRESS IN REACTION KINETICS AND MECHANISM 2019. [DOI: 10.3184/007967405779134029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The kinetics and mechanism of action of the most intensively studied mammalian peroxidases, myeloperoxidase and prostaglandin H synthase are critically reviewed. Evidence against currently favored mechanisms is presented. It is shown that myeloperoxidase has a strong defence mechanism against free hypochlorous acid, commonly thought to be its principal product in its bactericidal activity. Rather, after its two-electron oxidation of chloride ion, myeloperoxidase rapidly converts it into an enzyme-bound chlorinating intermediate, most likely a chlorinated distal imidazole ring. This species chlorinates taurine which may either be a transfer agent of Cl+ to other species or may act directly in attack on invading microorganisms. The currently favored mechanism of action of prostaglandin H synthase-1 is a branching chain mechanism in which Compound I is converted into a species containing a tyrosyl radical on the opposite side of the enzyme. Once the tyrosyl radical is formed it converts arachidonic acid into a peroxide in a cyclooxygenase reaction, independent of the peroxidase activity. This mechanism cannot explain the enhancing effect of small free radical scavengers, nor the fact that peroxidase activity continues unabated while the cyclooxygenase reaction is proceeding, nor the 2: 1 ratio of small free radical scavenger to arachidonic acid consumption. A tightly coupling of peroxidase and cycloxygenase reactions appears to be the steady state mechanism, and the branching chain mechanism, if it occurs, is confined to a burst transient state phase.
Collapse
Affiliation(s)
- H. Brian Dunford
- Emeritus Professor of Biophysical Chemistry, Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2 Current address: 1119 Pecan Lake Court, Stillwater, OK, USA 74074-1181 Phone/Fax 405-624-3322
| |
Collapse
|
16
|
Yoshida E, Kurita M, Eto K, Kumagai Y, Kaji T. Methylmercury promotes prostacyclin release from cultured human brain microvascular endothelial cells via induction of cyclooxygenase-2 through activation of the EGFR-p38 MAPK pathway by inhibiting protein tyrosine phosphatase 1B activity. Toxicology 2017; 392:40-46. [PMID: 28958600 DOI: 10.1016/j.tox.2017.09.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/22/2017] [Accepted: 09/24/2017] [Indexed: 12/26/2022]
Abstract
Methylmercury is an environmental pollutant that exhibits neurotoxicity when ingested, primarily in the form of neuropathological lesions that localize along deep sulci and fissures, in addition to edematous and inflammatory changes in patient cerebrums. These conditions been known to give rise to a variety of ailments that have come to be collectively termed Minamata disease. Since prostaglandins I2 and E2 (PGI2 and PGE2) increase vascular permeability and contribute to the progression of inflammatory changes, we hypothesize that methylmercury induces the synthesis of these prostaglandins in brain microvascular endothelial cells and pericytes. To test this theory, human brain microvascular endothelial cells and pericytes were cultured and treated with methylmercury, after which the PGI2 and PGE2 released from endothelial cells and/or pericytes were quantified by enzyme-linked immunosorbent assay while protein and mRNA expressions in endothelial cells were analyzed by western blot analysis and real-time reverse transcription polymerase chain reaction, respectively. Experimental results indicate that methylmercury inhibits the activity of protein tyrosine phosphatase 1B, which in turn activates the epidermal growth factor receptor-p38 mitogen-activated protein kinase pathway that induces cyclooxygenase-2 expression. It was also found that the cyclic adenosine 3',5'-monophosphate pathway, which can be activated by PGI2 and PGE2, is involved in methylmercury-induced cyclooxygenase-2 expression. Since it appears that protein tyrosine phosphatase 1 B serves as a sensor protein for methylmercury in these mechanisms, it is our belief that the results of the present study may provide additional insights into the molecular mechanisms responsible for edematous and inflammatory changes in the cerebrum of patients with Minamata disease.
Collapse
Affiliation(s)
- Eiko Yoshida
- Department of Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda 278-8510, Japan
| | - Masaru Kurita
- Department of Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda 278-8510, Japan
| | - Komyo Eto
- Health and Nursing Facilities for the Aged, Jushindai, Shinwakai, 272 Ikura Kitakata, Tamana 865-0041, Japan
| | - Yoshito Kumagai
- Environmental Biology Section, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Japan
| | - Toshiyuki Kaji
- Department of Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda 278-8510, Japan.
| |
Collapse
|
17
|
Seo MJ, Oh DK. Prostaglandin synthases: Molecular characterization and involvement in prostaglandin biosynthesis. Prog Lipid Res 2017; 66:50-68. [DOI: 10.1016/j.plipres.2017.04.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 03/30/2017] [Accepted: 04/01/2017] [Indexed: 01/30/2023]
|
18
|
|
19
|
Aljuhani N, Whittal RM, Khan SR, Siraki AG. Phenylbutazone Oxidation via Cu,Zn-SOD Peroxidase Activity: An EPR Study. Chem Res Toxicol 2015; 28:1476-83. [DOI: 10.1021/acs.chemrestox.5b00152] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Naif Aljuhani
- Pharmacology
and Toxicology Department, Faculty of Pharmacy, Taibah University, Madinah, Saudi Arabia
| | | | | | | |
Collapse
|
20
|
Stepensky D, Rimon G. Competition between low-dose aspirin and other NSAIDs for COX-1 binding and its clinical consequences for the drugs’ antiplatelet effects. Expert Opin Drug Metab Toxicol 2014; 11:41-52. [DOI: 10.1517/17425255.2014.971010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- David Stepensky
- Ben-Gurion University of the Negev, Department of Clinical Biochemistry and Pharmacology, The Faculty of Health Sciences, P.O.Box 653, Beer Sheva 84105, Israel
| | - Gilad Rimon
- Ben-Gurion University of the Negev, Department of Clinical Biochemistry and Pharmacology, The Faculty of Health Sciences, P.O.Box 653, Beer Sheva 84105, Israel
| |
Collapse
|
21
|
Why do a wide variety of animals retain multiple isoforms of cyclooxygenase? Prostaglandins Other Lipid Mediat 2014; 109-111:14-22. [PMID: 24721150 DOI: 10.1016/j.prostaglandins.2014.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 03/10/2014] [Accepted: 03/26/2014] [Indexed: 12/14/2022]
Abstract
Cyclooxygenase (COX) has been cloned from the phyla Cnidaria, Mollusca, Arthropoda, and Chordata of the animal kingdom. Many organisms have multiple COX isoforms that have arisen from gene duplication. It is not well understood why there are multiple COX isoforms in the same organism, or when duplication of the COX gene occurred. Here, we summarize the current knowledge of the evolutionary history of COX in the animal kingdom and discuss the reasons why the multiple COX system has been retained so widely. The phylogenetic analysis suggests that all COX genes in animals may descend from a common ancestor and that the duplication of an ancestral COX gene might occur within each lineage after the divergence of the animal. In most instances, the expressions of multiple COX isoforms are separately regulated and these isoforms play different and important pathophysiological roles in each organism. This may be the reason why multiple COX isoforms are widely retained.
Collapse
|
22
|
Kim SF. The Nitric Oxide-Mediated Regulation of Prostaglandin Signaling in Medicine. VITAMINS & HORMONES 2014; 96:211-45. [DOI: 10.1016/b978-0-12-800254-4.00009-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
23
|
Salvemini D, Kim SF, Mollace V. Reciprocal regulation of the nitric oxide and cyclooxygenase pathway in pathophysiology: relevance and clinical implications. Am J Physiol Regul Integr Comp Physiol 2013; 304:R473-87. [PMID: 23389111 DOI: 10.1152/ajpregu.00355.2012] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The nitric oxide (NO) and cyclooxygenase (COX) pathways share a number of similarities. Nitric oxide is the mediator generated from the NO synthase (NOS) pathway, and COX converts arachidonic acid to prostaglandins, prostacyclin, and thromboxane A(2). Two major forms of NOS and COX have been identified to date. The constitutive isoforms critically regulate several physiological states. The inducible isoforms are overexpressed during inflammation in a variety of cells, producing large amounts of NO and prostaglandins, which may underlie pathological processes. The cross-talk between the COX and NOS pathways was initially reported by Salvemini and colleagues in 1993, when they demonstrated in a series of in vitro and in vivo studies that NO activates the COX enzymes to produce increased amounts of prostaglandins. Those studies led to the concept that COX enzymes represent important endogenous "receptor" targets for amplifying or modulating the multifaceted roles of NO in physiology and pathology. Since then, numerous studies have furthered our mechanistic understanding of these interactions in pathophysiological settings and delineated potential clinical outcomes. In addition, emerging evidence suggests that the canonical nitroxidative species (NO, superoxide, and/or peroxynitrite) modulate biosynthesis of prostaglandins through non-COX-related pathways. This article provides a comprehensive state-of-the art overview in this area.
Collapse
Affiliation(s)
- Daniela Salvemini
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, MO 63104, USA.
| | | | | |
Collapse
|
24
|
Malhotra S, Deshmukh SS, Dastidar SG. COX inhibitors for airway inflammation. Expert Opin Ther Targets 2012; 16:195-207. [PMID: 22324934 DOI: 10.1517/14728222.2012.661416] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION The cyclooxygenase (COX) enzyme, which is responsible for the production of prostaglandins (PGs), key mediators of inflammation, may have the potential to become an attractive target for anti-inflammatory therapy. COX catalyzes the conversion of arachidonic acid (AA) into PGs, which play a significant role in disease. PGs are lipid mediators of central importance in the regulation of inflammation and smooth muscle tone. Airway-resident inflammatory cells release PGs: PGD2 and PDF2a amplify smooth muscle contraction and airway inflammation. Following its conversion from membrane phospholipids by phospholipase, AA enters the prostanoid pathway via COX, which catalyzes the conversion of AA to PGH2. PGH2 is then converted to biologically active PGs by cell-specific PG synthases. As COX is the rate limiting step in the PG pathway, the regulation of this enzyme is of critical importance in PG production. AREAS COVERED This review addresses the opportunities and challenges of COX inhibitors as therapeutic targets in airway inflammation. The review covers literature from the past 20 years. EXPERT OPINION Current literature favors COX inhibitors as potential targets for airway diseases. However, from the information available, it is not clear whether the COX enzyme by itself can serve as a target in drug development for asthma and COPD. Therefore, additional research is required to elucidate the mechanisms of action of COX metabolites before it can be considered as a target.
Collapse
Affiliation(s)
- Sanjay Malhotra
- Daiichi Sankyo India Pharma Private Ltd., Department of Chemistry, Haryana, India.
| | | | | |
Collapse
|
25
|
Purification and characterization of the recombinant human prostaglandin H synthase-2 expressed in Pichia pastoris. Protein Expr Purif 2012; 83:182-9. [DOI: 10.1016/j.pep.2012.03.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 03/28/2012] [Accepted: 03/29/2012] [Indexed: 11/17/2022]
|
26
|
Barroso-Neto IL, Marques JPC, da Costa RF, Caetano EWS, Cavada BS, Gottfried C, Freire VN. Inactivation of Ovine Cyclooxygenase-1 by Bromoaspirin and Aspirin: A Quantum Chemistry Description. J Phys Chem B 2012; 116:3270-9. [DOI: 10.1021/jp206397z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Ito L. Barroso-Neto
- Department of Biochemistry, Universidade Federal do Ceará, Fortaleza 60455-760, Ceará, Brazil
| | - João Paulo C. Marques
- Department of Physics, Universidade Federal do Ceará, Fortaleza 60455-760,
Ceará, Brazil
| | - Roner F. da Costa
- Department of Physics, Universidade Federal do Ceará, Fortaleza 60455-760,
Ceará, Brazil
| | - Ewerton W. S. Caetano
- Instituto
Federal de Educação, Ciência e Tecnologia do Ceará, Fortaleza 60040-531, Ceará,
Brazil
| | - Benildo S. Cavada
- Department of Biochemistry, Universidade Federal do Ceará, Fortaleza 60455-760, Ceará, Brazil
| | - Carmem Gottfried
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre
90035-003, Rio Grande do Sul, Brazil
| | - Valder N. Freire
- Department of Physics, Universidade Federal do Ceará, Fortaleza 60455-760,
Ceará, Brazil
| |
Collapse
|
27
|
Kaur J, Bhardwaj A, Huang Z, Knaus EE. N-1 and C-3 substituted indole Schiff bases as selective COX-2 inhibitors: synthesis and biological evaluation. Bioorg Med Chem Lett 2012; 22:2154-9. [PMID: 22361134 DOI: 10.1016/j.bmcl.2012.01.130] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 01/25/2012] [Accepted: 01/30/2012] [Indexed: 10/14/2022]
Abstract
A group of N-1 and C-3 disubstituted-indole Schiff bases bearing an indole N-1 (R'=H, CH(2)Ph, COPh) substituent in conjunction with a C-3 -C=HN-C(6)H(4)-4-X (X=F, Me, CF(3), Cl) substituent were synthesized and evaluated as inhibitors of cyclooxygenase (COX) isozymes (COX-1/COX-2). Within this group of Schiff bases, compounds 15 (R(1)=CH(2)Ph, X=F), 17 (R(1)=CH(2)Ph, X=CF(3)), 18 (R(1)=COPh, X=F) and 20 (R(1)=COPh, X=CF(3)) were identified as effective and selective COX-2 inhibitors (COX-2 IC(50)'s=0.32-0.84 μM range; COX-2 selectivity index (SI)=113 to >312 range). 1-Benzoyl-3-[(4-trifluoromethylphenylimino)methyl]indole (20) emerged as the most potent (COX-1 IC(50) >100 μM; COX-2 IC(50)=0.32 μM) and selective (SI >312) COX-2 inhibitor. Furthermore, compound 20 is a selective COX-2 inhibitor in contrast to the reference drug indomethacin that is a potent and selective COX-1 inhibitor (COX-1 IC(50)=0.13 μM; COX-2 IC(50)=6.9 μM, COX-2 SI=0.02). Molecular modeling studies employing compound 20 showed that the phenyl CF(3) substituent attached to the CN spacer is positioned near the secondary pocket of the COX-2 active site, the CN nitrogen atom is hydrogen bonded (N···NH=2.85 Å) to the H90 residue, and the indole N-1 benzoyl is positioned in a hydrophobic pocket of the COX-2 active site near W387.
Collapse
Affiliation(s)
- Jatinder Kaur
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | |
Collapse
|
28
|
Sevigny MB, Graham K, Ponce E, Louie MC, Mitchell K. Glycosylation of human cyclooxygenase-2 (COX-2) decreases the efficacy of certain COX-2 inhibitors. Pharmacol Res 2012; 65:445-50. [PMID: 22245433 DOI: 10.1016/j.phrs.2012.01.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2011] [Revised: 12/30/2011] [Accepted: 01/01/2012] [Indexed: 12/15/2022]
Abstract
Prostanoids play an important role in a variety of physiological and pathophysiological processes including inflammation and cancer. The rate-limiting step in the prostanoid biosynthesis pathway is catalyzed by cyclooxygenase-2 (COX-2). COX-2 exists as two glycoforms, 72 and 74 kDa, the latter resulting from an additional glycosylation at Asn(580). In this study, Asn(580) was mutated, and the mutant and wild-type COX-2 genes were expressed in COS-1 cells to determine how glycosylation affects the inhibition of COX-2 activity by aspirin, flurbiprofen, ibuprofen, celecoxib, and etoricoxib. Results indicate that certain inhibitors were 2-5 times more effective at inhibiting COX-2 activity when the glycosylation site was eliminated, indicating that glycosylation of COX-2 at Asn(580) decreases the efficacy of some inhibitors.
Collapse
Affiliation(s)
- Mary B Sevigny
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael, CA 94901, USA.
| | | | | | | | | |
Collapse
|
29
|
Abstract
Chagas disease is caused by Trypanosoma cruzi, a protozoan parasite. Chagas disease remains a serious health problem in large parts of Mexico and Central and South America, where it is a major cause of morbidity and mortality. This disease is being increasingly recognized in non-endemic regions due to immigration. Heart disease develops in 10-30% of infected individuals. It is increasingly clear that parasite- and host-derived bioactive lipids potently modulate disease progression. Many of the changes that occur during acute and chronic Chagas disease can be accounted for by the effects of arachidonic acid (AA)-derived lipids such as leukotrienes, lipoxins, H(P)ETEs, prostaglandins (PGs) and thromboxane. During the course of infection with T. cruzi, changes in circulating levels of AA metabolites are observed. Antagonism of PG synthesis with cyclooxygenase (COX) inhibitors has both beneficial and adverse effects. Treatment with COX inhibitors during acute infection may result in increased parasite load and mortality. However, treatment instituted during chronic infection may be beneficial with no increase in mortality and substantial improvement with cardiac function. Recently, T. cruzi infection of mice deficient in AA biosynthetic enzymes for various pathways has yielded more insightful data than pharmacological inhibition and has highlighted the potential deleterious effects of inhibitors due to "off-target" actions. Using COX-1 null mice, it was observed that parasite biosynthesis is dependent upon host metabolism, that the majority of TXA(2) liberated during T. cruzi infection is derived from the parasite and that this molecule may act as a quorum sensor to control parasite growth/differentiation. Thus, eicosanoids present during acute infection may act as immunomodulators aiding the transition to, and maintenance of, the chronic stage of the disease. It is also likely that the same mediators that initially function to ensure host survival may later contribute to cardiovascular damage. Collectively, the eicosanoids represent a new series of targets for therapy in Chagas disease with defined potential therapeutic windows in which to apply these agents for greatest effect. A deeper understanding of the mechanism of action of non-steroidal anti-inflammatory drugs may provide clues to the differences between host responses in acute and chronic T. cruzi infection.
Collapse
|
30
|
Kaur J, Bhardwaj A, Huang Z, Knaus EE. Aspirin analogues as dual cyclooxygenase-2/5-lipoxygenase inhibitors: synthesis, nitric oxide release, molecular modeling, and biological evaluation as anti-inflammatory agents. ChemMedChem 2011; 7:144-50. [PMID: 22095955 DOI: 10.1002/cmdc.201100460] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Indexed: 01/02/2023]
Abstract
Analogues of aspirin were synthesized through an efficient one-step reaction in which the carboxyl group was replaced by an ethyl ester, and/or the acetoxy group was replaced by an N-substituted sulfonamide (SO(2)NHOR(2):R(2) =H, Me, CH(2)Ph) pharmacophore. These analogues were designed for evaluation as dual cyclooxygenase-2 (COX-2) and 5-lipoxygenase (5-LOX) inhibitors. In vitro COX-1/COX-2 isozyme inhibition studies identified compounds 11 (CO(2) H, SO(2)NHOH), 12 (CO(2)H, SO(2)NHOCH(2)Ph), and 16 (CO(2)Et, SO(2)NHOH) as highly potent and selective COX-2 inhibitors (IC(50) range: 0.07-0.7 μM), which exhibited appreciable in vivo anti-inflammatory activity (ED(50) range: 23.1-31.4 mg kg(-1)). Moreover, compounds 11 (IC(50) =0.2 μM) and 16 (IC(50) =0.3 μM), with a sulfohydroxamic acid (SO(2)NHOH) moiety showed potent 5-LOX inhibitory activity. Furthermore, the SO(2)NHOH moiety present in compounds 11 and 16 was found to be a good nitric oxide (NO) donor upon incubation in phosphate buffer at pH 7.4. Molecular docking studies in the active binding site of COX-2 and 5-LOX provided complementary theoretical support for the experimental biological structure-activity data acquired.
Collapse
Affiliation(s)
- Jatinder Kaur
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | | | | | | |
Collapse
|
31
|
Aspirin: pharmacology and clinical applications. THROMBOSIS 2011; 2012:173124. [PMID: 22195279 PMCID: PMC3236360 DOI: 10.1155/2012/173124] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 10/03/2011] [Indexed: 12/21/2022]
Abstract
Antiplatelet therapy has been documented to reduce risks of cardiovascular disease after acute myocardial infarction, coronary artery bypass graft, and in chronic atrial fibrillation patients, amongst other risk factors. Conventional management of thrombosis-based disorders includes the use of heparin, oral anticoagulants, and the preferred antiplatelet agent aspirin. Interestingly, aspirin was not intended to be used as an antiplatelet agent; rather, after being repurposed, it has become one of the most widely prescribed antithrombotic drugs. To this end, there have been several milestones in the development of antiplatelet agents in the last few decades, such as adenosine diphosphate receptor inhibitors, phosphodiesterase inhibitors, and GPIIb/IIIa inhibitors. However, given some of the limitations of these therapies, aspirin continues to play a major role in the management of thrombotic and cardiovascular disorders and is expected to do so for years to come.
Collapse
|
32
|
Kim SF. The role of nitric oxide in prostaglandin biology; update. Nitric Oxide 2011; 25:255-64. [PMID: 21820072 DOI: 10.1016/j.niox.2011.07.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Revised: 07/15/2011] [Accepted: 07/19/2011] [Indexed: 02/06/2023]
Abstract
The biosynthesis of nitric oxide (NO) and prostaglandin share many similarities. Two major forms of nitric oxide synthase (NOS) and cyclooxygenase (COX) have been identified: constitutive versus inducible. In general, the constitutive form functions in housekeeping and physiologic roles whereas the inducible form is up-regulated by mitogenic or inflammatory stimuli and is responsible for pathophysiological responses. The cross talk between the COX and NOS pathways was initially reported in 1993 and since then, numerous studies have been undertaken to delineate the functional consequences of this interaction as well as the potential mechanism by which each pathway interacts. This review will focus in particular on recent advances in this field that extend our understanding of these two pathways under various systems.
Collapse
Affiliation(s)
- Sangwon F Kim
- Department of Psychiatry and Pharmacology, Center for Neurobiology and Behavior, University of Pennsylvania School of Medicine, 125 S, 31st St. TRL Rm 2207, Philadelphia, PA 19104, USA.
| |
Collapse
|
33
|
Miura D, Miura Y, Yagasaki K. Restoration by Prostaglandins E(2) and F (2) (alpha) of Resveratrol-Induced Suppression of Hepatoma Cell Invasion in Culture. Cytotechnology 2011; 43:155-9. [PMID: 19003221 DOI: 10.1023/b:cyto.0000039903.22449.79] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In our previous study, resveratrol, a polyphenolic compound in grapes with antioxidative property, and resveratol-loaded rat serum (RS) were found to suppress the invasion of AH109A cells, an ascite hepatoma cell line. The aim of the present study was to investigate whether and which prostaglandins (PGs) would be involved in the invasion of AH109A cells and its suppression by resveratrol and resveratrol-loaded RS, using an in vitro invasion assay system. Not only PGE(2) but also PGF(2) (alpha) stimulated the spontaneous invasion of AH109A cells.They also canceled the resveratrol-induced suppression of hepatoma cell invasion. Results obtained suggest an involvement of PGs, especially PGE(2), in the invasion of hepatoma cells.
Collapse
Affiliation(s)
- Daiki Miura
- Department of Applied Biological Science, Tokyo Noko University, Saiwaicho 3-5-8, Fuchu, Tokyo, 183-8509, Japan
| | | | | |
Collapse
|
34
|
Roongsitthichai A, Srisuwatanasagul S, Koonjaenak S, Tummaruk P. Expression of cyclooxygenase-2 in the endometrium of gilts with different stages of endometritis. J Vet Med Sci 2011; 73:1425-31. [PMID: 21720106 DOI: 10.1292/jvms.11-0130] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The present study determined the association among the expression of COX-2, stages of endometritis, and types and number of local immune cells infiltrating into the gilts' endometrium. The uterine tissues from 24 Landrace x Yorkshire gilts identified as acute endometritis (n = 7), chronic endometritis (n = 7), and normal endometrium (n = 10) were included. The tissues were prepared for both histological and immunohistochemical investigations. The immunoexpression of COX-2 in every layer of the gilts' endometria was appraised by avidin-biotin-peroxidase complex method via image analysis; and was reported as percentage of positive area and staining index. The results revealed that the immunoexpression of COX-2 was found only in the surface epithelial layer. The gilts with acute endometritis possessed higher both percentage of positive area (68.99% versus 4.50% and 3.43%, P < 0.001) and staining index (1.13 versus 0.05 and 0.04, P < 0.001) than those with chronic endometritis and normal endometrium, respectively. Positive correlations between the number of surface epithelial neutrophils and percentage of COX-2 positive area (r = 0.47, P = 0.022), as well as mean staining index (r = 0.44, P = 0.032) were observed. In conclusion, the immunoexpression of COX-2 was found strongest in the gilts with acute endometritis, meanwhile it was not different between those with chronic endometritis and normal endometrium. This suggested that the expression of COX-2 might be dependent not only on the infiltration of local immune cells in the endometrium, but also on the duration of exposure with inflammatory agents.
Collapse
Affiliation(s)
- Atthaporn Roongsitthichai
- Departments of Obstetrics, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | | | | | | |
Collapse
|
35
|
Trushkin NA, Filimonov IS, Vrzheshch PV. Inhibition of cyclooxygenase activity of prostaglandin-H-synthase by excess substrate (molecular oxygen). BIOCHEMISTRY (MOSCOW) 2011; 75:1368-73. [PMID: 21314604 DOI: 10.1134/s0006297910110088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
For the cyclooxygenase reaction of prostaglandin-H-synthase isolated from ram vesicular glands, dependences of the initial reaction rate, the maximal yield of the product, and the rate constant of enzyme inactivation in the course of reaction on oxygen concentration were studied in the absence and in the presence of electron donor in the reaction medium. It is shown that in the absence of electron donor the cyclooxygenase reaction is strictly governed by Michaelis-Menten kinetics over a wide range of oxygen concentrations (5-800 µM). In the presence of electron donor in the reaction medium it was found that cyclooxygenase reaction is inhibited by an excess of dissolved oxygen: the maximal values of the initial reaction rate and yield of the product are attained at oxygen concentration 50 µM, and its increase to 500 µM causes twofold decrease in the initial rate and maximal yield. The rate constant of enzyme inactivation in the course of reaction increases on increase in oxygen concentration both in the presence and in the absence of electron donor.
Collapse
Affiliation(s)
- N A Trushkin
- International Research Center for Biochemical Technology, Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119992, Russia
| | | | | |
Collapse
|
36
|
Helicobacter pylori and gastric cancer: factors that modulate disease risk. Clin Microbiol Rev 2010; 23:713-39. [PMID: 20930071 DOI: 10.1128/cmr.00011-10] [Citation(s) in RCA: 993] [Impact Index Per Article: 66.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Helicobacter pylori is a gastric pathogen that colonizes approximately 50% of the world's population. Infection with H. pylori causes chronic inflammation and significantly increases the risk of developing duodenal and gastric ulcer disease and gastric cancer. Infection with H. pylori is the strongest known risk factor for gastric cancer, which is the second leading cause of cancer-related deaths worldwide. Once H. pylori colonizes the gastric environment, it persists for the lifetime of the host, suggesting that the host immune response is ineffective in clearing this bacterium. In this review, we discuss the host immune response and examine other host factors that increase the pathogenic potential of this bacterium, including host polymorphisms, alterations to the apical-junctional complex, and the effects of environmental factors. In addition to host effects and responses, H. pylori strains are genetically diverse. We discuss the main virulence determinants in H. pylori strains and the correlation between these and the diverse clinical outcomes following H. pylori infection. Since H. pylori inhibits the gastric epithelium of half of the world, it is crucial that we continue to gain understanding of host and microbial factors that increase the risk of developing more severe clinical outcomes.
Collapse
|
37
|
Lee JW. [Treatment update on portal hypertension and complications]. THE KOREAN JOURNAL OF GASTROENTEROLOGY 2010; 56:144-54. [PMID: 20847605 DOI: 10.4166/kjg.2010.56.3.144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Current understanding of the pathophysiology of portal hypertension has resulted in therapeutic approaches aimed at correcting the increased splanchnic blood flow and some of which have been already used in clinical practice. Recently new perspectives opened and erstwhile paradigm has been changed to focus on increased resistance to portal blood flow and the formation of portosystemic collateralization. Several studies revealed the clear-cut mechanisms of hepatic endothelial dysfunction and abnormal angiogenesis contributing to the development of portal hypertension. Thus the modulations of hyperdynamic circulation or angiogenesis seem to be valuable therapeutic targets. In the current review update, we discuss the multidisciplinary management of modulating hepatic vascular resistance and abnormal angiogenesis associated with portal hypertension. However, these new pharmacological approaches are still under investigation and widescale clinical application are needed to develop effective strategies.
Collapse
Affiliation(s)
- Jin Woo Lee
- Department of Internal Medicine, Inha University School of Medicine, Incheon, Korea.
| |
Collapse
|
38
|
Bosch J, Abraldes JG, Fernández M, García-Pagán JC. Hepatic endothelial dysfunction and abnormal angiogenesis: new targets in the treatment of portal hypertension. J Hepatol 2010; 53:558-67. [PMID: 20561700 DOI: 10.1016/j.jhep.2010.03.021] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Revised: 03/19/2010] [Accepted: 03/19/2010] [Indexed: 12/12/2022]
Abstract
Portal hypertension is the main cause of complications in patients with chronic liver disease. Over the past 25 years, progress in the understanding of the pathophysiology of portal hypertension was followed by the introduction of an effective pharmacological therapy, consisting mainly of treatments aimed at correcting the increased splanchnic blood flow. It is only recently that this paradigm has been changed. Progress in our knowledge of the mechanisms of increased resistance to portal blood flow, of the formation of portal-systemic collaterals, and of mechanisms other than vasodilatation maintaining the increased splanchnic blood flow have opened entirely new perspectives for developing more effective treatment strategies. This is the aim of the current review, which focuses on: (a) the modulation of hepatic vascular resistance by correcting the increased hepatic vascular tone due to hepatic endothelial dysfunction, and (b) correcting the abnormal angiogenesis associated with portal hypertension, which contributes to liver inflammation and fibrogenesis, to the hyperkinetic splanchnic circulation, and to the formation of portal-systemic collaterals and varices.
Collapse
Affiliation(s)
- Jaume Bosch
- Hepatic Hemodynamic Laboratory, Liver Unit, Hospital Clínic-IDIBAPS and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Spain.
| | | | | | | |
Collapse
|
39
|
Sonzogni-Desautels K, Knapp DW, Sartin E, Doré M. Effect of cyclooxygenase inhibitors in a xenograft model of canine mammary tumours. Vet Comp Oncol 2010; 9:161-71. [PMID: 21848619 DOI: 10.1111/j.1476-5829.2010.00242.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Inhibition of cyclooxygenase-2 (COX-2) represents a possible avenue for the prevention and/or treatment of some cancers. Our goal was to compare the effect of a selective inhibitor of COX-2, deracoxib, and a COX-1 and -2 inhibitor, piroxicam, on the growth of canine mammary tumours in a murine model. CMT-9 was used to induce xenografts in nude mice. Mice were treated with piroxicam (0.6 mg kg(-1)), deracoxib (6 mg kg(-1)) or a control solution. Tumour volumes between 0 and 24 days post-treatment showed no significant difference between all groups. A second series of experiments was performed with a higher dose of piroxicam (0.9 mg kg(-1)). Tumour volumes between 14 and 21 days post-treatment were significantly smaller in piroxicam-treated mice compared with controls. These results demonstrate that COX inhibition reduced the growth of canine mammary cancer xenografts in mice, suggesting that COX inhibitors could have a positive effect in dogs.
Collapse
Affiliation(s)
- K Sonzogni-Desautels
- Faculté de médecine vétérinaire, Département de pathologie et microbiologie, Université de Montréal, 3200 Sicotte, Saint-Hyacinthe, Québec, Canada
| | | | | | | |
Collapse
|
40
|
Structural basis for certain naturally occurring bioflavonoids to function as reducing co-substrates of cyclooxygenase I and II. PLoS One 2010; 5:e12316. [PMID: 20808785 PMCID: PMC2925883 DOI: 10.1371/journal.pone.0012316] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Accepted: 07/26/2010] [Indexed: 02/07/2023] Open
Abstract
Background Recent studies showed that some of the dietary bioflavonoids can strongly stimulate the catalytic activity of cyclooxygenase (COX) I and II in vitro and in vivo, presumably by facilitating enzyme re-activation. In this study, we sought to understand the structural basis of COX activation by these dietary compounds. Methodology/Principal Findings A combination of molecular modeling studies, biochemical analysis and site-directed mutagenesis assay was used as research tools. Three-dimensional quantitative structure-activity relationship analysis (QSAR/CoMFA) predicted that the ability of bioflavonoids to activate COX I and II depends heavily on their B-ring structure, a moiety known to be associated with strong antioxidant ability. Using the homology modeling and docking approaches, we identified the peroxidase active site of COX I and II as the binding site for bioflavonoids. Upon binding to this site, bioflavonoid can directly interact with hematin of the COX enzyme and facilitate the electron transfer from bioflavonoid to hematin. The docking results were verified by biochemical analysis, which reveals that when the cyclooxygenase activity of COXs is inhibited by covalent modification, myricetin can still stimulate the conversion of PGG2 to PGE2, a reaction selectively catalyzed by the peroxidase activity. Using the site-directed mutagenesis analysis, we confirmed that Q189 at the peroxidase site of COX II is essential for bioflavonoids to bind and re-activate its catalytic activity. Conclusions/Significance These findings provide the structural basis for bioflavonoids to function as high-affinity reducing co-substrates of COXs through binding to the peroxidase active site, facilitating electron transfer and enzyme re-activation.
Collapse
|
41
|
Botting RM. Vane’s discovery of the mechanism of action of aspirin changed our understanding of its clinical pharmacology. Pharmacol Rep 2010; 62:518-25. [DOI: 10.1016/s1734-1140(10)70308-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Revised: 05/01/2010] [Indexed: 01/08/2023]
|
42
|
Menter DG, Schilsky RL, DuBois RN. Cyclooxygenase-2 and cancer treatment: understanding the risk should be worth the reward. Clin Cancer Res 2010; 16:1384-90. [PMID: 20179228 PMCID: PMC4307592 DOI: 10.1158/1078-0432.ccr-09-0788] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Targeting the prostaglandin (PG) pathway is potentially a critical intervention for the prevention and treatment of cancer. Central to PG biosynthesis are two isoforms of cyclooxygenase (COX 1 and 2), which produce prostaglandin H(2) (PGH(2)) from plasma membrane stores of fatty acids. COX-1 is constitutively expressed, whereas COX-2 is an inducible isoform upregulated in many cancers. Differences between COX-1 and COX-2 catalytic sites enabled development of selective inhibitors. Downstream of the COX enzymes, prostaglandin E(2) synthase converts available PGH(2) to prostaglandin E(2) (PGE(2)), which can stimulate cancer progression. Significant research efforts are helping identify more selective targets and fully elucidate the downstream targets of prostaglandin E(2)-mediated oncogenesis. Nonetheless, as a key rate-limiting control point of PG biosynthesis, COX-2 continues to be an important anticancer target. As we embark upon a new era of individualized medicine, a better understanding of the individual risk and/or benefit involved in COX-2 selective targeting is rapidly evolving. This review endeavors to summarize developments in our understanding of COX-2 and its downstream targets as vital areas of anticancer research and to provide the current status of an exciting aspect of molecular medicine.
Collapse
Affiliation(s)
- David G. Menter
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | | | - Raymond N. DuBois
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
- Department of GI Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| |
Collapse
|
43
|
Wu G, Tsai AL, Kulmacz RJ. Cyclooxygenase competitive inhibitors alter tyrosyl radical dynamics in prostaglandin H synthase-2. Biochemistry 2010; 48:11902-11. [PMID: 19894761 DOI: 10.1021/bi901600f] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Reaction of prostaglandin H synthase (PGHS) isoforms 1 or 2 with peroxide forms a radical at Tyr385 that is required for cyclooxygenase catalysis and another radical at Tyr504, whose function is unknown. Both tyrosyl radicals are transient and rapidly dissipated by reductants, suggesting that cyclooxygenase catalysis might be vulnerable to suppression by intracellular antioxidants. Our initial hypothesis was that the two radicals are in equilibrium and that their proportions and stability are altered upon binding of fatty acid substrate. As a test, we examined the effects of three competitive inhibitors (nimesulide, flurbiprofen, and diclofenac) on the proportions and stability of the two radicals in PGHS-2 pretreated with peroxide. Adding nimesulide after ethyl peroxide led to some narrowing of the tyrosyl radical signal detected by EPR spectroscopy, consistent with a small increase in the proportion of the Tyr504 radical. Neither flurbiprofen nor diclofenac changed the EPR line width when added after peroxide. In contrast, the effects of cyclooxygenase inhibitors on the stability of the preformed tyrosyl radicals were dramatic. The half-life of total tyrosyl radical was 4.1 min in the control, >10 h with added nimesulide, 48 min with flurbiprofen, and 0.8 min with diclofenac. Stabilization of the tyrosyl radicals was evident even at substoichiometric levels of nimesulide. Thus, the inhibitors had potent, structure-dependent, effects on the stability of both tyrosyl radicals. This dramatic modulation of tyrosyl radical stability by cyclooxygenase site ligands suggests a mechanism for regulating the reactivity of PGHS tyrosyl radicals with cellular antioxidants.
Collapse
Affiliation(s)
- Gang Wu
- Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | | | | |
Collapse
|
44
|
Bai HW, Zhu BT. Myricetin and quercetin are naturally occurring co-substrates of cyclooxygenases in vivo. Prostaglandins Leukot Essent Fatty Acids 2010; 82:45-50. [PMID: 19897347 PMCID: PMC2818348 DOI: 10.1016/j.plefa.2009.10.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Accepted: 10/13/2009] [Indexed: 12/25/2022]
Abstract
Bioflavonoids are ubiquitously present in the plant kingdom, and some of them are presently being sold as healthy dietary supplements around the world. Recently, it was shown that some of the dietary polyphenols were strong stimulators of the catalytic activity of cyclooxygenase I and II, resulting in increased formation of certain prostaglandin (PG) products in vitro and also in intact cells in culture. In the present study, we investigated the effect of two representative dietary compounds, quercetin and myricetin, on plasma and tissue levels of several PG products in normal Sprague-Dawley rats. We found that these two dietary bioflavonoids could strongly stimulate the formation of PG products in vivo in a time-dependent manner, and the stimulatory effect of these two bioflavonoids was dose-dependent with a unique biphasic pattern. At lower doses (<0.3mg/kg b.w.), they strongly stimulated the formation of PGE(2), but at higher doses (>0.3mg/kg b.w.), there was a dose-dependent reduction of the stimulatory effect. These results provide support for the hypothesis that some of the bioflavonoids are naturally occurring physiological co-substrates for the cyclooxygenases in vivo.
Collapse
Affiliation(s)
- Hyoung-Woo Bai
- Department of Pharmacology, Toxicology and Therapeutics, School of Medicine, University of Kansas Medical Center, MS-1018, room KLSIC-4061, 2146 W. 39th Ave, Kansas City, KS 66160, USA
| | | |
Collapse
|
45
|
Tsai AL, Kulmacz RJ. Prostaglandin H synthase: resolved and unresolved mechanistic issues. Arch Biochem Biophys 2010; 493:103-24. [PMID: 19728984 PMCID: PMC2812681 DOI: 10.1016/j.abb.2009.08.019] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Revised: 08/25/2009] [Accepted: 08/26/2009] [Indexed: 11/20/2022]
Abstract
The cyclooxygenase and peroxidase activities of prostaglandin H synthase (PGHS)-1 and -2 have complex kinetics, with the cyclooxygenase exhibiting feedback activation by product peroxide and irreversible self-inactivation, and the peroxidase undergoing an independent self-inactivation process. The mechanistic bases for these complex, non-linear steady-state kinetics have been gradually elucidated by a combination of structure/function, spectroscopic and transient kinetic analyses. It is now apparent that most aspects of PGHS-1 and -2 catalysis can be accounted for by a branched chain radical mechanism involving a classic heme-based peroxidase cycle and a radical-based cyclooxygenase cycle. The two cycles are linked by the Tyr385 radical, which originates from an oxidized peroxidase intermediate and begins the cyclooxygenase cycle by abstracting a hydrogen atom from the fatty acid substrate. Peroxidase cycle intermediates have been well characterized, and peroxidase self-inactivation has been kinetically linked to a damaging side reaction involving the oxyferryl heme oxidant in an intermediate that also contains the Tyr385 radical. The cyclooxygenase cycle intermediates are poorly characterized, with the exception of the Tyr385 radical and the initial arachidonate radical, which has a pentadiene structure involving C11-C15 of the fatty acid. Oxygen isotope effect studies suggest that formation of the arachidonate radical is reversible, a conclusion consistent with electron paramagnetic resonance spectroscopic observations, radical trapping by NO, and thermodynamic calculations, although moderate isotope selectivity was found for the H-abstraction step as well. Reaction with peroxide also produces an alternate radical at Tyr504 that is linked to cyclooxygenase activation efficiency and may serve as a reservoir of oxidizing equivalent. The interconversions among radicals on Tyr385, on Tyr504, and on arachidonate, and their relationships to regulation and inactivation of the cyclooxygenase, are still under active investigation for both PGHS isozymes.
Collapse
Affiliation(s)
- Ah-Lim Tsai
- Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA.
| | | |
Collapse
|
46
|
Turman MV, Marnett LJ. Prostaglandin Endoperoxide Synthases. COMPREHENSIVE NATURAL PRODUCTS II 2010:35-63. [DOI: 10.1016/b978-008045382-8.00028-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
47
|
Feitoza CQ, Semedo P, Gonçalves GM, Cenedeze MA, Pinheiro HS, Dos Santos OFP, Landgraf RG, Pacheco-Silva A, Câmara NOS. Modulation of inflammatory response by selective inhibition of cyclooxygenase-1 and cyclooxygenase-2 in acute kidney injury. Inflamm Res 2009; 59:167-75. [PMID: 19711010 DOI: 10.1007/s00011-009-0083-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Revised: 08/07/2009] [Accepted: 08/10/2009] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE AND DESIGN This work explored the role of inhibition of cyclooxygenases (COXs) in modulating the inflammatory response triggered by acute kidney injury. MATERIAL C57Bl/6 mice were used. TREATMENT Animals were treated or not with indomethacin (IMT) prior to injury (days -1 and 0). METHODS Animals were subjected to 45 min of renal pedicle occlusion and sacrificed at 24 h after reperfusion. Serum creatinine and blood urea nitrogen, reactive oxygen species (ROS), kidney myeloperoxidase (MPO) activity, and prostaglandin E2 (PGE(2)) levels were analyzed. Tumor necrosis factor (TNF)-alpha, t-bet, interleukin (IL)-10, IL-1beta, heme oxygenase (HO)-1, and prostaglandin E synthase (PGES) messenger RNA (mRNA) were studied. Cytokines were quantified in serum. RESULTS IMT-treated animals presented better renal function with less acute tubular necrosis and reduced ROS and MPO production. Moreover, the treatment was associated with lower expression of TNF-alpha, PGE(2), PGES, and t-bet and upregulation of HO-1 and IL-10. This profile was mirrored in serum, where inhibition of COXs significantly decreased interferon (IFN)-gamma, TNF-alpha, and IL-12 p70 and upregulated IL-10. CONCLUSIONS COXs seem to play an important role in renal ischemia and reperfusion injury, involving the secretion of pro-inflammatory cytokines, activation of neutrophils, and ROS production. Inhibition of COX pathway is intrinsically involved with cytoprotection.
Collapse
Affiliation(s)
- Carla Q Feitoza
- Laboratory of Experimental and Clinical Immunology, Nephrology Division, Federal University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Alcohol dehydrogenase (ADH) was used as a marker molecule to clarify the mechanism of gastric mucosal damage as a side effect of using piroxicam. Piroxicam inactivated ADH during interaction of ADH with horseradish peroxidase and H2O2 (HRP-H2O2). The ADH was more easily inactivated under aerobic than anaerobic conditions, indicating participation by oxygen. Superoxide dismutase, but not hydroxyl radical scavengers, inhibited inactivation of ADH, indicating participation by superoxide. Sulfhydryl (SH) groups in ADH were lost during incubation of piroxicam with HRP-H2O2. Adding reduced glutathione (GSH) efficiently blocked ADH inactivation. Other SH enzymes, including creatine kinase and glyceraldehyde-3-phosphate dehydrogenase, were also inactivated by piroxicam with HRP-H2O2. Thus SH groups in the enzymes seem vulnerable to piroxicam activated by HRP-H2O2. Spectral change in piroxicam was caused by HRP-H2O2. ESR signals of glutathionyl radicals occurred during incubation of piroxicam with HRP-H2O2 in the presence of GSH. Under anaerobic conditions, glutathionyl radical formation increased. Thus piroxicam free radicals interact with GSH to produce glutathionyl radicals. Piroxicam peroxyl radicals or superoxide, or both, seem to inactivate ADH. Superoxide may be produced through interaction of peroxyl radicals with H2O2. Thus superoxide dismutase may inhibit inactivation of ADH through reducing piroxicam peroxyl radicals or blocking interaction of SH groups with O2 , or both. Other oxicam derivatives, including isoxicam, tenoxicam and meloxicam, induced ADH inactivation in the presence of HRP-H2O2.
Collapse
Affiliation(s)
- Sanae Muraoka
- Department of Biology, Hokkaido College of Pharmacy, Katsuraoka-cho 7-1, Otaru 0470264, Japan
| | | |
Collapse
|
49
|
Czembirek C, Eder-Czembirek C, Erovic BM, Turhani D, Spittler A, Selzer E, Pötter R, Thurnher D. The cyclooxygenase-2 inhibitor nimesulide, a nonsteroidal analgesic, decreases the effect of radiation therapy in head-and-neck cancer cells. Strahlenther Onkol 2009; 185:310-7. [PMID: 19440670 DOI: 10.1007/s00066-009-1929-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2008] [Accepted: 11/17/2008] [Indexed: 10/20/2022]
Abstract
BACKGROUND No data are available on the effects of the cyclooxygenase-2 (COX-2) inhibitor nimesulide in combination with irradiation on the survival of head-and-neck carcinoma cells. MATERIAL AND METHODS Two head-and-neck carcinoma cell lines (SCC9 and SCC25) were treated with nimesulide (50-600 microM) and irradiated concomitantly or sequentially. Early effects on cell survival were investigated by counting cell numbers, long-term effects by colony-forming assays. Cell-cycle effects were analyzed 24-72 h after treatment with nimesulide by flow cytometry. RESULTS Unexpectedly, nimesulide solely inhibited cell proliferation without affecting colony-forming ability. In addition, no evidence for a radiosensitizing effect of nimesulide in short-term assays was seen. Nimesulide alone had no effect on clonogenic survival alone or in combination with radiation. CONCLUSION Nimesulide differentially affects cell proliferation and clonogenic survival and may decrease the efficacy of radiotherapy. Short-term assays to assess tumor growth may not correctly predict the clinically relevant long-term effect of COX-2 inhibitors.
Collapse
Affiliation(s)
- Cornelia Czembirek
- Department of Cranio-, Maxillofacial and Oral Surgery, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Kawakami Y, Nakamura T, Hosokawa T, Suzuki-Yamamoto T, Yamashita H, Kimoto M, Tsuji H, Yoshida H, Hada T, Takahashi Y. Antiproliferative activity of guava leaf extract via inhibition of prostaglandin endoperoxide H synthase isoforms. Prostaglandins Leukot Essent Fatty Acids 2009; 80:239-45. [PMID: 19457650 DOI: 10.1016/j.plefa.2009.04.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2008] [Revised: 04/03/2009] [Accepted: 04/23/2009] [Indexed: 11/20/2022]
Abstract
Prostaglandin endoperoxide H synthase (PGHS) is a key enzyme for the synthesis of prostaglandins (PGs) which play important roles in inflammation and carcinogenesis. Because the extract from Psidium guajava is known to have a variety of beneficial effects on our body including the anti-inflammatory, antioxidative and antiproliferative activities, we investigated whether the extract inhibited the catalytic activity of the two PGHS isoforms using linoleic acid as an alternative substrate. The guava leaf extract inhibited the cyclooxygenase reaction of recombinant human PGHS-1 and PGHS-2 as assessed by conversion of linoleic acid to 9- and 13-hydroxyoctadecadienoic acids (HODEs). The guava leaf extract also inhibited the PG hydroperoxidase activity of PGHS-1, which was not affected by nonsteroidal anti-inflammatory drugs (NSAIDs). Quercetin which was one of the major components not only inhibited the cyclooxygenase activity of both isoforms but also partially inhibited the PG hydroperoxidase activity. Overexpression of human PGHS-1 and PGHS-2 in the human colon carcinoma cells increased the DNA synthesis rate as compared with mock-transfected cells which did not express any isoforms. The guava leaf extract not only inhibited the PGE(2) synthesis but also suppressed the DNA synthesis rate in the PGHS-1- and PGHS-2-expressing cells to the same level as mock-transfected cells. These results demonstrate the antiproliferative activity of the guava leaf extract which is at least in part caused by inhibition of the catalytic activity of PGHS isoforms.
Collapse
Affiliation(s)
- Yuki Kawakami
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama 719-1197, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|