1
|
Zhao Q, Dai W, Chen HY, Jacobs RE, Zlokovic BV, Lund BT, Montagne A, Bonnin A. Prenatal disruption of blood-brain barrier formation via cyclooxygenase activation leads to lifelong brain inflammation. Proc Natl Acad Sci U S A 2022; 119:e2113310119. [PMID: 35377817 PMCID: PMC9169666 DOI: 10.1073/pnas.2113310119] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 02/14/2022] [Indexed: 11/21/2022] Open
Abstract
Gestational maternal immune activation (MIA) in mice induces persistent brain microglial activation and a range of neuropathologies in the adult offspring. Although long-term phenotypes are well documented, how MIA in utero leads to persistent brain inflammation is not well understood. Here, we found that offspring of mothers treated with polyriboinosinic–polyribocytidylic acid [poly(I:C)] to induce MIA at gestational day 13 exhibit blood–brain barrier (BBB) dysfunction throughout life. Live MRI in utero revealed fetal BBB hyperpermeability 2 d after MIA. Decreased pericyte–endothelium coupling in cerebral blood vessels and increased microglial activation were found in fetal and 1- and 6-mo-old offspring brains. The long-lasting disruptions result from abnormal prenatal BBB formation, driven by increased proliferation of cyclooxygenase-2 (COX2; Ptgs2)-expressing microglia in fetal brain parenchyma and perivascular spaces. Targeted deletion of the Ptgs2 gene in fetal myeloid cells or treatment with the inhibitor celecoxib 24 h after immune activation prevented microglial proliferation and disruption of BBB formation and function, showing that prenatal COX2 activation is a causal pathway of MIA effects. Thus, gestational MIA disrupts fetal BBB formation, inducing persistent BBB dysfunction, which promotes microglial overactivation and behavioral alterations across the offspring life span. Taken together, the data suggest that gestational MIA disruption of BBB formation could be an etiological contributor to neuropsychiatric disorders.
Collapse
Affiliation(s)
- Qiuying Zhao
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
| | - Weiye Dai
- Master of Science, Molecular Pharmacology and Toxicology, School of Pharmacy, University of Southern California, Los Angeles, CA 90089
| | - Hui Yu Chen
- Master of Medical Physiology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
| | - Russell E. Jacobs
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
| | - Berislav V. Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
| | - Brett T. Lund
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
| | - Axel Montagne
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, EH16 4SB Edinburgh, United Kingdom
- Centre for Clinical Brain Sciences, Edinburgh BioQuarter, EH16 4SB Edinburgh, United Kingdom
| | - Alexandre Bonnin
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
| |
Collapse
|
2
|
Jougleux JL, Rioux FM, Fiset S, Boudreau LH, Surette ME. Influence of iron-deficient diets during gestation and lactation on cerebral fatty acids and eicosanoids in guinea pig offspring-Comparison of studies with different sources of dietary lipids. Prostaglandins Leukot Essent Fatty Acids 2019; 149:37-45. [PMID: 31422159 DOI: 10.1016/j.plefa.2019.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/26/2019] [Accepted: 08/07/2019] [Indexed: 11/29/2022]
Abstract
Previous studies showed that mild iron deficiency anaemia (IDA) induced by feeding an iron deficient (ID) diet to female guinea pigs during gestation and lactation to alters the auditory functions of the offspring when corn oil is the only source of dietary lipids. Conversely, feeding an ID diet with a dietary fatty acid composition similar to that of typical human western diets induced minor impairments. Since tissue fatty acid metabolism is affected by dietary iron, the current study measured the impacts of these ID diets (ID-corn and ID-west) compared to the corresponding iron-sufficient control diets (IS-corn and IS-west) on encephalum fatty acid metabolism in the offspring at post-natal day 24. IDA induced by the ID-corn diet resulted in significant increases in encephalum n-6 PUFA content, but IDA induced by the ID-west diet had little impact on fatty acid profiles compared to the IS-west group. Brain COX II protein expression and FADS2 mRNA expression were statistically unaffected in both experiments, but encephalum PGE2 concentrations were significantly reduced in ID-west pups. These results suggest IDA studies during prenatal development should consider dietary lipid compositions.
Collapse
Affiliation(s)
- Jean-Luc Jougleux
- Département de Chimie et Biochimie, Université de Moncton, 18 Antonine-Maillet Avenue, Moncton, NB, E1A 3E9, Canada
| | - France M Rioux
- École des sciences de la Nutrition, Faculté des Sciences de la Santé, Université d'Ottawa, Ottawa, ON, K1N 6N5, Canada
| | - Sylvain Fiset
- Secteur Administration et Sciences Humaines, Université de Moncton, Campus Edmundston, Edmundston, NB, E3V 2S8, Canada
| | - Luc H Boudreau
- Département de Chimie et Biochimie, Université de Moncton, 18 Antonine-Maillet Avenue, Moncton, NB, E1A 3E9, Canada
| | - Marc E Surette
- Département de Chimie et Biochimie, Université de Moncton, 18 Antonine-Maillet Avenue, Moncton, NB, E1A 3E9, Canada.
| |
Collapse
|
3
|
Ahmad AA, Randall MD, Roberts RE. Sex differences in the role of phospholipase A 2 -dependent arachidonic acid pathway in the perivascular adipose tissue function in pigs. J Physiol 2017; 595:6623-6634. [PMID: 28877347 DOI: 10.1113/jp274831] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 08/21/2017] [Indexed: 12/18/2022] Open
Abstract
KEY POINTS The fat surrounding blood vessels (perivascular adipose tissue or PVAT) releases vasoactive compounds that regulate vascular smooth muscle tone. There are sex differences in the regulation of vascular tone, but, to date, no study has investigated whether there are sex differences in the regulation of blood vessel tone by PVAT. This study has identified that the cyclooxygenase products thromboxane and PGF2α are released from coronary artery PVAT from pigs. Thromboxane appears to mediate the PVAT-induced contraction in arteries from females, whereas PGF2α appears to mediate the contraction in arteries from males. These sex differences in the role of these prostanoids in the PVAT-induced contraction can be explained by a greater release of thromboxane from PVAT from female animals and greater sensitivity to PGF2α in the porcine coronary artery from males. ABSTRACT Previous studies have demonstrated that perivascular adipose tissue (PVAT) causes vasoconstriction. In this present study, we determined the role of cyclooxygenase-derived prostanoids in this contractile response and determined whether there were any sex differences in the regulation of vascular tone by PVAT. Contractions in isolated segments of coronary arteries were determined using isolated tissue baths and isometric tension recording. Segments were initially cleaned of PVAT, which was then re-added to the tissue bath and changes in tone measured over 1 h. Levels of PGF2α and thromboxane B2 (TXB2 ) were quantified by ELISA, and PGF2α (FP) and thromboxane A2 (TP) receptor expression determined by Western blotting. In arteries from both male and female pigs, re-addition of PVAT caused a contraction, which was partially inhibited by the cyclooxygenase inhibitors indomethacin and flurbiprofen. The FP receptor antagonist AL8810 attenuated the PVAT-induced contraction in arteries from males, whereas the TP receptor antagonist GR32191B inhibited the PVAT-induced contraction in arteries from females. Although there was no difference in PGF2α levels in PVAT between females and males, PGF2α produced a larger contraction in arteries from males, correlating with a higher FP receptor expression. In contrast, release of TXB2 from PVAT from females was greater than from males, but there was no difference in the contraction by the TXA2 agonist U46619, or TP receptor expression in arteries from different sexes. These findings demonstrate clear sex differences in PVAT function in which PGF2α and TXA2 antagonists can inhibit the PVAT-induced vasoconstriction in male and female PCAs, respectively.
Collapse
Affiliation(s)
- Abdulla A Ahmad
- Cell Signalling and Pharmacology Research Group, School of Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Michael D Randall
- Cell Signalling and Pharmacology Research Group, School of Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Richard E Roberts
- Cell Signalling and Pharmacology Research Group, School of Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| |
Collapse
|
4
|
Lee JC. Therapeutic effect of prostaglandin E1 in monocrotaline-induced pulmonary arterial hypertension rats. Anat Cell Biol 2017; 50:60-68. [PMID: 28417056 PMCID: PMC5386927 DOI: 10.5115/acb.2017.50.1.60] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/06/2017] [Accepted: 03/08/2017] [Indexed: 02/08/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe pulmonary vascular disease characterized by sustained increase in pulmonary arterial pressure and excessive thickening and remodeling of distal small pulmonary arteries. During disease progression, PAH include increase in mean pulmonary arterial pressure, right ventricular (RV) enlargement, increased pulmonary vascular resistance, and smooth muscle hypertrophy in pulmonary arterioles. Several anti-PAH therapies targeting various pathways involved in PAH progression have been approved by the Food and Drug Adminstration. However, many of the currently available anti-PAH drugs suffer from a number of limitations, including short biological half-life, and poor pulmonary selectivity. Prostaglandin E1 (PGE1) is a potent vasodilator with selectivity toward pulmonary circulation when it is administered via the pulmonary route. However, PGE1 has a very short half-life of 5–10 minutes. Therefore, we hypothesized that long-term effect of PGE1 could reduce mal-adaptive structural remodeling of the lung and heart and prevent ventricular arrhythmias in monocrotaline-induced rat model of PAH. Our results revealed that PGE1 reduced ventricular hypertrophy, protein expressions of endothelin-1 and endothelin receptor A, and the expression of fibrosis. These results support the notion that PGE1 can improve the functional properties of RV, highlighting its potential benefits for heart and lung impairment.
Collapse
Affiliation(s)
- Jae Chul Lee
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Anatomy, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
5
|
Uekawa K, Koizumi K, Hwang J, Brunier N, Hattori Y, Zhou P, Park L. Obligatory Role of EP1 Receptors in the Increase in Cerebral Blood Flow Produced by Hypercapnia in the Mice. PLoS One 2016; 11:e0163329. [PMID: 27657726 PMCID: PMC5033465 DOI: 10.1371/journal.pone.0163329] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/07/2016] [Indexed: 12/11/2022] Open
Abstract
Hypercapnia induces potent vasodilation in the cerebral circulation. Although it has long been known that prostanoids participate in the cerebrovascular effects of hypercapnia, the role of prostaglandin E2 (PGE2) and PGE2 receptors have not been fully investigated. In this study, we sought to determine whether cyclooxygenase-1 (COX-1)-derived PGE2 and EP1 receptors are involved in the cerebrovascular response induced by hypercapnia. Cerebral blood flow (CBF) was recorded by laser-Doppler flowmetry in the somatosenasory cortex of anesthetized male EP1-/- mice and wild type (WT) littermates. In WT mice, neocortical application of the EP1 receptor antagonist SC-51089 attenuated the increase in CBF elicited by systemic hypercapnia (pCO2 = 50-60 mmHg). SC-51089 also attenuated the increase in CBF produced by neocortical treatment of arachidonic acid or PGE2. These CBF responses were also attenuated in EP1-/- mice. In WT mice, the COX-1 inhibitor SC-560, but not the COX-2 inhibitor NS-398, attenuated the hypercapnic CBF increase. Neocortical application of exogenous PGE2 restored the attenuation in resting CBF and the hypercapnic response induced by SC-560. In contrast, exogenous PGE2 failed to rescue the attenuation both in WT mice induced by SC-51089 and EP1-/- mice, attesting to the obligatory role of EP1 receptors in the response. These findings indicate that the hypercapnic vasodilatation depends on COX-1-derived PGE2 acting on EP1 receptors and highlight the critical role that COX-1-derived PGE2 and EP1 receptors play in the hypercapnic regulation of the cerebral circulation.
Collapse
Affiliation(s)
- Ken Uekawa
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, 10065, United States of America
| | - Kenzo Koizumi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, 10065, United States of America
| | - Jason Hwang
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, 10065, United States of America
| | - Nathalie Brunier
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, 10065, United States of America
| | - Yorito Hattori
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, 10065, United States of America
| | - Ping Zhou
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, 10065, United States of America
| | - Laibaik Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, 10065, United States of America
- * E-mail:
| |
Collapse
|
6
|
Kraemer MP, Choi H, Reese J, Lamb FS, Breyer RM. Regulation of arterial reactivity by concurrent signaling through the E-prostanoid receptor 3 and angiotensin receptor 1. Vascul Pharmacol 2016; 84:47-54. [PMID: 27260940 DOI: 10.1016/j.vph.2016.05.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 05/20/2016] [Accepted: 05/30/2016] [Indexed: 12/28/2022]
Abstract
Prostaglandin E2 (PGE2), a cyclooxygenase metabolite that generally acts as a systemic vasodepressor, has been shown to have vasopressor effects under certain physiologic conditions. Previous studies have demonstrated that PGE2 receptor signaling modulates angiotensin II (Ang II)-induced hypertension, but the interaction of these two systems in the regulation of vascular reactivity is incompletely characterized. We hypothesized that Ang II, a principal effector of the renin-angiotensin-aldosterone system, potentiates PGE2-mediated vasoconstriction. Here we demonstrate that pre-treatment of arterial rings with 1nM Ang II potentiated PGE2-evoked constriction in a concentration dependent manner (AUC-Ang II 2.778±2.091, AUC+Ang II 22.830±8.560, ***P<0.001). Using genetic deletion models and pharmacological antagonists, we demonstrate that this potentiation effect is mediated via concurrent signaling between the angiotensin II receptor 1 (AT1) and the PGE2 E-prostanoid receptor 3 (EP3) in the mouse femoral artery. EP3 receptor-mediated vasoconstriction is shown to be dependent on extracellular calcium in combination with proline-rich tyrosine kinase 2 (Pyk2) and Rho-kinase. Thus, our findings reveal a novel mechanism through which Ang II and PGE2 regulate peripheral vascular reactivity.
Collapse
Affiliation(s)
- Maria P Kraemer
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN, United States.
| | - Hyehun Choi
- Department of Pediatrics, Division of Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jeff Reese
- Department of Pediatrics, Division of Neonatology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Fred S Lamb
- Department of Pediatrics, Division of Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Richard M Breyer
- Department of Medicine, Veterans Affairs Hospital, Nashville, TN, United States; Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN, United States; Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
7
|
Baretella O, Vanhoutte P. Endothelium-Dependent Contractions. ADVANCES IN PHARMACOLOGY 2016; 77:177-208. [DOI: 10.1016/bs.apha.2016.04.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
8
|
Dabertrand F, Hannah RM, Pearson JM, Hill-Eubanks DC, Brayden JE, Nelson MT. Prostaglandin E2, a postulated astrocyte-derived neurovascular coupling agent, constricts rather than dilates parenchymal arterioles. J Cereb Blood Flow Metab 2013; 33:479-82. [PMID: 23385200 PMCID: PMC3618402 DOI: 10.1038/jcbfm.2013.9] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
It has been proposed that prostaglandin E(2) (PGE(2)) is released from astrocytic endfeet to dilate parenchymal arterioles through activation of prostanoid (EP(4)) receptors during neurovascular coupling. However, the direct effects of PGE(2) on isolated parenchymal arterioles have not been tested. Here, we examined the effects of PGE(2) on the diameter of isolated pressurized parenchymal arterioles from rat and mouse brain. Contrary to the prevailing assumption, we found that PGE(2) (0.1, 1, and 5 μmol/L) constricted rather than dilated parenchymal arterioles. Vasoconstriction to PGE(2) was prevented by inhibitors of EP(1) receptors. These results strongly argue against a direct role of PGE(2) on arterioles during neurovascular coupling.
Collapse
Affiliation(s)
- Fabrice Dabertrand
- Department of Pharmacology, College of Medicine, University of Vermont, Burlington, VT 05405-0068, USA
| | | | | | | | | | | |
Collapse
|
9
|
Neurogenic contractions in intraocular porcine ciliary arteries are mediated by α2-adrenoceptors and NPY1 receptors and are inhibited by prostaglandin E2 acting on prejunctional EP4 receptors. Exp Eye Res 2013. [DOI: 10.1016/j.exer.2012.11.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
10
|
Tayman C, Rayyan M, Allegaert K. Neonatal pharmacology: extensive interindividual variability despite limited size. J Pediatr Pharmacol Ther 2012; 16:170-84. [PMID: 22479159 DOI: 10.5863/1551-6776-16.3.170] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Providing safe and effective drug therapy to neonates requires knowledge of the impact of development on the pharmacokinetics and pharmacodynamics of drugs. Although maturational changes are observed throughout childhood, they are most prominent during the first year of life. Several of these processes overlap, making development an extremely dynamic system in the newborn compared with that in infants, children, or adults. Changes in body composition and porportions, liver mass, metabolic activity, and renal function collectively affect the pharmacokinetic behavior of medications. Instead of simply adapting doses by scaling adult or pediatric doses on the basis of a patient's weight and/or body surface area, integrated knowledge of clinical maturation and developmental pharmacology is critical to the safe and effective use of medications in neonates. Unfortunately, the effects of human ontogeny on both pharmacokinetics and pharmacodynamics have not been well established in these early stages of life, and information regarding the influence of developmental changes on the pharmacodynamics of medications is even more limited. Theoretically, age-dependent variations in receptor number and affinity for drugs have significant potential to influence an individual's response to drug therapy. In this review, some of the relevant covariates of pharmacokinetics and pharmacodynamics in neonates are reviewed and illustrated based on the published literature.
Collapse
|
11
|
Majed BH, Khalil RA. Molecular mechanisms regulating the vascular prostacyclin pathways and their adaptation during pregnancy and in the newborn. Pharmacol Rev 2012; 64:540-82. [PMID: 22679221 DOI: 10.1124/pr.111.004770] [Citation(s) in RCA: 173] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Prostacyclin (PGI(2)) is a member of the prostanoid group of eicosanoids that regulate homeostasis, hemostasis, smooth muscle function and inflammation. Prostanoids are derived from arachidonic acid by the sequential actions of phospholipase A(2), cyclooxygenase (COX), and specific prostaglandin (PG) synthases. There are two major COX enzymes, COX1 and COX2, that differ in structure, tissue distribution, subcellular localization, and function. COX1 is largely constitutively expressed, whereas COX2 is induced at sites of inflammation and vascular injury. PGI(2) is produced by endothelial cells and influences many cardiovascular processes. PGI(2) acts mainly on the prostacyclin (IP) receptor, but because of receptor homology, PGI(2) analogs such as iloprost may act on other prostanoid receptors with variable affinities. PGI(2)/IP interaction stimulates G protein-coupled increase in cAMP and protein kinase A, resulting in decreased [Ca(2+)](i), and could also cause inhibition of Rho kinase, leading to vascular smooth muscle relaxation. In addition, PGI(2) intracrine signaling may target nuclear peroxisome proliferator-activated receptors and regulate gene transcription. PGI(2) counteracts the vasoconstrictor and platelet aggregation effects of thromboxane A(2) (TXA(2)), and both prostanoids create an important balance in cardiovascular homeostasis. The PGI(2)/TXA(2) balance is particularly critical in the regulation of maternal and fetal vascular function during pregnancy and in the newborn. A decrease in PGI(2)/TXA(2) ratio in the maternal, fetal, and neonatal circulation may contribute to preeclampsia, intrauterine growth restriction, and persistent pulmonary hypertension of the newborn (PPHN), respectively. On the other hand, increased PGI(2) activity may contribute to patent ductus arteriosus (PDA) and intraventricular hemorrhage in premature newborns. These observations have raised interest in the use of COX inhibitors and PGI(2) analogs in the management of pregnancy-associated and neonatal vascular disorders. The use of aspirin to decrease TXA(2) synthesis has shown little benefit in preeclampsia, whereas indomethacin and ibuprofen are used effectively to close PDA in the premature newborn. PGI(2) analogs have been used effectively in primary pulmonary hypertension in adults and have shown promise in PPHN. Careful examination of PGI(2) metabolism and the complex interplay with other prostanoids will help design specific modulators of the PGI(2)-dependent pathways for the management of pregnancy-related and neonatal vascular disorders.
Collapse
Affiliation(s)
- Batoule H Majed
- Harvard Medical School, Brigham and Women's Hospital, Division of Vascular Surgery, 75 Francis St., Boston, MA 02115, USA
| | | |
Collapse
|
12
|
Dasgupta E, Singh G. Vaginal Misoprostol vs Vaginal Misoprostol With Estradiol for Labor Induction: A Prospective Double Blind Study. J Obstet Gynaecol India 2012; 62:47-51. [PMID: 23372290 DOI: 10.1007/s13224-012-0156-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Accepted: 02/15/2011] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE To compare the safety and effectiveness of vaginal misoprostol with combined vaginal misoprostol and estradiol for induction of labor in unfavorable cervix. METHOD A prospective study was carried out from Jan 2008 to Jul 2008 on total of 90 women with unfavorable cervix (Bishop's score was <5) and gestation >36 weeks with clinical indication for induction of labor. They were randomly assigned to receive either vaginal misoprostol 25 μg alone or vaginal misoprostol 25 μg with vaginal estradiol 50 μg. Misoprostol alone was repeated every 3 h in both groups till ripening of cervix (Bishop's score was = 8) and establishment of active labor. RESULTS Main indications were post dated pregnancies (period of gestation >41 weeks) and pregnancy induced hypertension. Age, parity and mode of delivery were not significantly different. No significant difference was found in pre induction Bishop's score, fetal outcome and maternal complications. However, doses of misoprostol required for cervical ripening (p = 0.017), time required for cervical ripening (p = 0.042), time required for starting of active labor (p = 0.017) and time required for delivery in vaginal delivery cases (p = 0.047) were found significantly less in combined estradiol and misoprostol group. CONCLUSION Estradiol acts synergistically with misoprostol vaginally and significantly hastens the process of cervical ripening, initiation of active labor and vaginal delivery.
Collapse
Affiliation(s)
- Ellora Dasgupta
- Department of Obstetrics & Gynecology, NC Jindal Institute of Medical Sciences, Model Town, Hisar 125005 India
| | | |
Collapse
|
13
|
Inflammatory Pathways in Parkinson's Disease; A BNE Microarray Study. PARKINSONS DISEASE 2012; 2012:214714. [PMID: 22548201 PMCID: PMC3324922 DOI: 10.1155/2012/214714] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2011] [Accepted: 01/04/2012] [Indexed: 12/18/2022]
Abstract
The aetiology of Parkinson's disease (PD) is yet to be fully understood but it is becoming more and more evident that neuronal cell death may be multifactorial in essence. The main focus of PD research is to better understand substantia nigra homeostasis disruption, particularly in relation to the wide-spread deposition of the aberrant protein α-synuclein. Microarray technology contributed towards PD research with several studies to date and one gene, ALDH1A1 (Aldehyde dehydrogenase 1 family, member A1), consistently reappeared across studies including the present study, highlighting dopamine (DA) metabolism dysfunction resulting in oxidative stress and most probably leading to neuronal cell death. Neuronal cell death leads to increased inflammation through the activation of astrocytes and microglia. Using our dataset, we aimed to isolate some of these pathways so to offer potential novel neuroprotective therapeutic avenues. To that effect our study has focused on the upregulation of P2X7 (purinergic receptor P2X, ligand-gated ion channel, 7) receptor pathway (microglial activation) and on the NOS3 (nitric oxide synthase 3) pathway (angiogenesis). In summary, although the exact initiator of striatal DA neuronal cell death remains to be determined, based on our analysis, this event does not remain without consequence. Extracellular ATP and reactive astrocytes appear to be responsible for the activation of microglia which in turn release proinflammatory cytokines contributing further to the parkinsonian condition. In addition to tackling oxidative stress pathways we also suggest to reduce microglial and endothelial activation to support neuronal outgrowth.
Collapse
|
14
|
Ishida K, Matsumoto T, Taguchi K, Kamata K, Kobayashi T. Protein kinase C delta contributes to increase in EP3 agonist-induced contraction in mesenteric arteries from type 2 diabetic Goto-Kakizaki rats. Pflugers Arch 2012; 463:593-602. [PMID: 22371141 DOI: 10.1007/s00424-012-1088-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 02/02/2012] [Accepted: 02/16/2012] [Indexed: 01/26/2023]
Abstract
Prostaglandin E(2) (PGE(2)), an important and ubiquitously present vasoactive eicosanoid, may either constrict or dilate systemic vascular beds. However, little is known about the vascular contractile responsiveness to and signaling pathways for PGE(2) at the chronic stage of type 2 diabetes. We hypothesized that PGE(2)-induced arterial contraction is augmented in type 2 diabetic Goto-Kakizaki (GK) rats via the protein kinase Cδ (PKCδ) pathway. Here, we investigated the vasoconstrictor effects of PGE(2) and of sulprostone (EP1-/EP3-receptor agonist) in rings cut from superior mesenteric arteries isolated from GK rats (37-44 weeks old). In arteries from GK rats (vs. those from age-matched Wistar rats), examined in the presence of a nitric oxide synthase inhibitor: 1) the PGE(2)- and sulprostone-induced vasocontractions (which were not blocked by the selective EP1 receptor antagonist sc19220) were enhanced, and these enhancements were suppressed by rottlerin (selective PKCδ inhibitor) but not by Gö6976 (selective PKCα/β inhibitor); 2) the sulprostone-stimulated phosphorylation of PKCδ (at Thr(505)), which yields an active form, was increased and 3) sulprostone-stimulated caldesmon phosphorylations, which are related to isometric force generation in smooth muscle, were increased. The protein expression of EP3 receptor in superior mesenteric arteries was similar between the two groups of rats. Our data suggest that the diabetes-related enhancement of EP3 receptor-mediated vasocontraction results from activation of the PKCδ pathway. Alterations in EP3 receptor-mediated vasocontraction may be important factors in the pathophysiological influences over arterial tone that are present in diabetic states.
Collapse
Affiliation(s)
- Keiko Ishida
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | | | | | | | | |
Collapse
|
15
|
Boutaud O, Roberts LJ. Mechanism-based therapeutic approaches to rhabdomyolysis-induced renal failure. Free Radic Biol Med 2011; 51:1062-7. [PMID: 21034813 PMCID: PMC3116013 DOI: 10.1016/j.freeradbiomed.2010.10.704] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 10/18/2010] [Accepted: 10/19/2010] [Indexed: 10/18/2022]
Abstract
Rhabdomyolysis-induced renal failure represents up to 15% of all cases of acute renal failure. Many studies over the past 4 decades have demonstrated that accumulation of myoglobin in the kidney is central in the mechanism leading to kidney injury. However, some discussion exists regarding the mechanism mediating this oxidant injury. Although the free-iron-catalyzed Fenton reaction has been proposed to explain the tissue injury, more recent evidence strongly suggests that the main cause of oxidant injury is myoglobin redox cycling and generation of oxidized lipids. These molecules can propagate tissue injury and cause renal vasoconstriction, two of the three main conditions associated with acute renal failure. This review presents the evidence supporting the two mechanisms of oxidative injury, describes the central role of myoglobin redox cycling in the pathology of renal failure associated with rhabdomyolysis, and discusses the value of therapeutic interventions aiming at inhibiting myoglobin redox cycling for the treatment of rhabdomyolysis-induced renal failure.
Collapse
Affiliation(s)
- Olivier Boutaud
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | | |
Collapse
|
16
|
van der Sterren S, Villamor E. Contractile effects of 15-E2t-isoprostane and 15-F2t-isoprostane on chicken embryo ductus arteriosus. Comp Biochem Physiol A Mol Integr Physiol 2011; 159:436-44. [PMID: 21565280 DOI: 10.1016/j.cbpa.2011.04.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Revised: 04/24/2011] [Accepted: 04/26/2011] [Indexed: 01/15/2023]
Abstract
Isoprostanes (IsoPs) are prostaglandin (PG)-like compounds produced nonenzymatically by free radical-catalyzed peroxidation of arachidonate. Cyclooxygenase-derived PGs play a major role in ductus arteriosus (DA) homeostasis but the putative role of IsoPs has not been studied so far. We investigated, using wire myography, the vasoactive effects of 15-E(2t)-IsoP and 15-F(2t)-IsoP in the chicken embryo DA, pulmonary artery (PA) and femoral artery (FA). 15-E(2t)-IsoP and 15-F(2t)-IsoP contracted DA, PA, and FA rings in a concentration-dependent manner. 15-E(2t)-IsoP was equally efficacious (mean±SE E(max)=1.25±0.06 mN/mm) as and more potent (-log of molar concentration producing 50% of E(max)=pEC(50)=7.00±0.04) than the thromboxane-prostanoid (TP) receptor agonist U46619 (E(max)=1.49±0.11 mN/mm; pEC(50)=6.48±0.05) in contracting chicken DA (pulmonary side). 15-F(2t)-IsoP was less potent (pEC(50)=5.74±0.11) and less efficacious (E(max)=0.96±0.11) than U46619. Concentration-dependent contractions to 15-E(2t)-IsoP and U46619 in DA rings were competitively inhibited by the TP receptor antagonist SQ29548 (0.1 μM to 10 μM) with no decrease in the E(max) values. SQ29548 also inhibited concentration-dependent contraction to 15-F(2t)-IsoP but this inhibition was associated with a decrease in E(max). Pre-incubation of DA rings with 15-F(2t)-IsoP inhibited responses to U46619 and, in vessels contracted with U46619 (1 μM), 15-F(2t)-IsoP (>1 μM) evoked a relaxant response. Enzyme immunoassay did not show a measurable release of 15-F(2t)-IsoP by DA rings. In conclusion, 15-E(2t)-IsoP is a potent and efficacious constrictor of chicken DA, acting through TP receptors. In contrast, 15-F(2t)-IsoP is probably acting as a partial agonist at TP receptors. We speculate that IsoPs play a role in the control of chicken DA tone and could participate in its closure.
Collapse
Affiliation(s)
- Saskia van der Sterren
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Developmental Biology (GROW), Maastricht, The Netherlands
| | | |
Collapse
|
17
|
Abstract
Understanding the role of ontogeny in the disposition and actions of medicines is the most fundamental prerequisite for safe and effective pharmacotherapeutics in the pediatric population. The maturational process represents a continuum of growth, differentiation, and development, which extends from the very small preterm newborn infant through childhood, adolescence, and to young adulthood. Developmental changes in physiology and, consequently, in pharmacology influence the efficacy, toxicity, and dosing regimen of medicines. Relevant periods of development are characterized by changes in body composition and proportion, developmental changes of physiology with pathophysiology, exposure to unique safety hazards, changes in drug disposition by major organs of metabolism and elimination, ontogeny of drug targets (e.g., enzymes, transporters, receptors, and channels), and environmental influences. These developmental components that result in critical windows of development of immature organ systems that may lead to permanent effects later in life interact in a complex, nonlinear fashion. The ontogeny of these physiologic processes provides the key to understanding the added dimension of development that defines the essential differences between children and adults. A basic understanding of the developmental dynamics in pediatric pharmacology is also essential to delineating the future directions and priority areas of pediatric drug research and development.
Collapse
MESH Headings
- Adolescent
- Body Composition/physiology
- Child
- Child, Preschool
- Drug-Related Side Effects and Adverse Reactions
- Female
- Human Development/physiology
- Humans
- Infant
- Infant, Newborn/physiology
- Infant, Newborn, Diseases/drug therapy
- Infant, Newborn, Diseases/physiopathology
- Infant, Premature/physiology
- Infant, Premature, Diseases/drug therapy
- Infant, Premature, Diseases/physiopathology
- Male
- Pediatrics
- Pharmaceutical Preparations/metabolism
- Pharmacokinetics
- Pharmacological Phenomena/physiology
Collapse
Affiliation(s)
- Hannsjörg W Seyberth
- Klinik fur Kinder- und Jugendmedizin, Philipps-Universität Marburg, Baldingerstraße, 35043 Marburg, Germany.
| | | |
Collapse
|
18
|
Antonucci R, Bassareo P, Zaffanello M, Pusceddu M, Fanos V. Patent ductus arteriosus in the preterm infant: new insights into pathogenesis and clinical management. J Matern Fetal Neonatal Med 2010; 23 Suppl 3:34-7. [DOI: 10.3109/14767058.2010.509920] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
19
|
|
20
|
Stimulation of prostaglandin EP2 receptors on RGC-5 cells in culture blunts the negative effect of serum withdrawal. Neurochem Res 2010; 35:820-9. [PMID: 20180018 DOI: 10.1007/s11064-010-0140-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2010] [Indexed: 10/19/2022]
Abstract
Reduced neurotrophic support is one possible cause for retinal ganglion cells dying in glaucoma. Experiments were designed to investigate the effect of EP2 receptor agonist butaprost on transformed retinal ganglion (RGC-5) cells where reduced neurotrophic support was simulated by serum withdrawal. Cultures were analysed for cell viability, flow cytometry, reactive oxygen species and apoptosis. Western blot and immunohistochemistry were used to provide information for the occurrence of PGE(2) receptor-types. We demonstrated the existence of all four types of PGE(2) receptors in RGC-5 cells and exposure of cultures to butaprost resulted in an elevation of cAMP. Serum deprivation induced RGC-5 cell death was significantly attenuated by butaprost as well as by rolipram and forskolin where intracellular cAMP levels were increased. These data are of value in relation to the possible use of EP2 receptor agonists to reduce both elevated intraocular pressure and retinal ganglion cell death as occurs in glaucoma.
Collapse
|
21
|
Fan FL, Zhu S, Chen LH, Zou YL, Fan LH, Kang JH, Ma AQ, Guan YF. Role of prostaglandin E and its receptors in the process of ductus arteriosus maturation and functional closure in the rabbit. Clin Exp Pharmacol Physiol 2010; 37:574-80. [PMID: 20082631 DOI: 10.1111/j.1440-1681.2010.05354.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
1. Patent ductus arteriosus (PDA) is a common congenital heart defect in premature infants. The present study was designed to determine the role of the prostaglandin (PG) E(2) pathway in the process of ductus arteriosus (DA) maturation and functional closure. 2. Changes in PGE(2) pathway-related enzymes and receptors in DA in preterm and term rabbits were examined at both the mRNA and protein levels. In addition, responses of DA rings to Po(2) and PGE(2) were determined. 3. Circulating PGE(2) levels remained high until 2 h after birth. High levels of the EP(4) receptor were found in preterm DA. These tissues were sensitive to PGE(2), which caused vessel dilation, but were insensitive to increased Po(2). In contrast, DA tissues from term rabbits exhibited an immediate contractile response to increased Po(2) and PGE(2) treatment resulted in vasoconstriction, which was associated with increased EP(3) and decreased EP(4) receptor expression in term DA. 4. In conclusion, the preterm PDA is maintained by high levels of PGE(2), which mainly binds to the EP(4) receptor under conditions of hypoxia. In contrast, in the term DA, in which levels of the EP(3) receptor are higher than in preterm DA, exposure to PGE(2) resulted in vasoconstriction under normoxic conditions. These findings suggest that blocking the EP(4) receptor may represent a more selective treatment for the preterm PDA, whereas activating the EP(3) receptor may be more suitable for the treatment of the term PDA.
Collapse
Affiliation(s)
- Fen-Ling Fan
- Department of Cardiovascular Medicine, The Key Laboratory of Environment and Gene-related Diseases, The Ministry of Education, Center of Cardiovascular Medicine of Xi'an Jiaotong University, Xi'an 710061, China
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Cąkała M, B. Strosznajder J. Znaczenie cyklooksygenaz w neurotoksyczności peptydów amyloidu β w chorobie Alzheimera. Neurol Neurochir Pol 2010; 44:65-79. [DOI: 10.1016/s0028-3843(14)60407-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
23
|
Burgoyne JR, Eaton P. Transnitrosylating nitric oxide species directly activate type I protein kinase A, providing a novel adenylate cyclase-independent cross-talk to beta-adrenergic-like signaling. J Biol Chem 2009; 284:29260-8. [PMID: 19726669 PMCID: PMC2785556 DOI: 10.1074/jbc.m109.046722] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Revised: 09/02/2009] [Indexed: 11/06/2022] Open
Abstract
The transnitrosylating nitric oxide (NO) donor nitrocysteine (CysNO) induced a disulfide bond between the two regulatory RI subunits of protein kinase A (PKA). The conventional NO donor S-nitroso-N-acetylpenicillamine failed to do this, consistent with our observation that it also did not promote protein S-nitrosylation. This disulfide oxidation event activated PKA and induced vasorelaxation independently of the classical beta-adrenergic or NO signaling pathway. Activation of PKA had also been anticipated to exert a positive inotropic effect on the myocardium but did not. The lack of positive inotropy was explained by CysNO concomitantly activating protein kinase G (PKG) Ialpha. PKG was found to exert a partial negative inotropic influence regardless of whether PKA was activated by classical beta-receptor stimulation or by disulfide bond formation. This work demonstrates that NO molecules that can induce S-nitrosylation directly activate type I PKA, providing a novel cross-talk to beta-adrenergic-like signaling without receptor or adenylate cyclase stimulation. However, the expected positive inotropic consequences of PKA activation by this novel mechanism are countermanded by the simultaneous dual activation of PKGIalpha, which is also activated by CysNO.
Collapse
Affiliation(s)
- Joseph R. Burgoyne
- From the Cardiovascular Division, The Rayne Institute, St. Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom
| | - Philip Eaton
- From the Cardiovascular Division, The Rayne Institute, St. Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom
| |
Collapse
|
24
|
Bonaventura D, Lunardi CN, Rodrigues GJ, Neto MA, Vercesi JA, de Lima RG, da Silva RS, Bendhack LM. Endothelium negatively modulates the vascular relaxation induced by nitric oxide donor, due to uncoupling NO synthase. J Inorg Biochem 2009; 103:1366-74. [DOI: 10.1016/j.jinorgbio.2009.07.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2009] [Revised: 07/22/2009] [Accepted: 07/24/2009] [Indexed: 10/20/2022]
|
25
|
Drug/nutrition interaction in the developing brain: Dipyrone enhances spreading depression in rats. Exp Neurol 2009; 219:492-8. [DOI: 10.1016/j.expneurol.2009.06.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Revised: 06/15/2009] [Accepted: 06/24/2009] [Indexed: 02/02/2023]
|
26
|
LeBlanc CP, Fiset S, Surette ME, Turgeon O'Brien H, Rioux FM. Maternal iron deficiency alters essential fatty acid and eicosanoid metabolism and increases locomotion in adult guinea pig offspring. J Nutr 2009; 139:1653-9. [PMID: 19640965 DOI: 10.3945/jn.109.106013] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Iron deficiency (ID) is the most prevalent worldwide nutritional deficiency. Groups at risk of developing ID anemia are infants and pregnant women, even in industrialized countries. Our goal in this study was to evaluate the long-term consequences of maternal ID on the offspring's fatty acid and eicosanoid metabolism, behavior, and spatial memory. Female guinea pigs consumed iron-sufficient (IS) and -deficient (ID) diets for 14 d before mating and throughout pregnancy and lactation. Dietary iron restriction resulted in ID in pregnant females. On postnatal d 9, all offspring (ID and IS) were weaned to the IS diet and at 42 d, all offspring were iron replete. Locomotion was tested in pups on postnatal d 24 and 40 and spatial memory from d 25 to 40. Pups from the ID group were significantly more active in the open field at both times of testing, whereas spatial memory, tested in a Morris water maze, was comparable in both groups. On postnatal d 42, liver, RBC, and brain fatty acid composition were measured. Dihomogammalinolenic [20:3(n-6)], docosapentaenoic [22:5(n-3)], and docosahexaenoic [22:6(n-3)] acid contents were significantly higher in brain phospholipids of offspring born to ID dams. Prostaglandin E(2) and F(2alpha) concentrations were also significantly higher in brains of offspring born to ID dams. This demonstrates that moderate ID during gestation and lactation results in alterations of brain fatty acid and eicosanoid metabolism and perturbation in behavior in adult offspring.
Collapse
Affiliation(s)
- Caroline P LeBlanc
- Ecole des sciences des aliments, de nutrition et d'études familiales, Université de Moncton, Moncton, NB E1A 3E9 Canada
| | | | | | | | | |
Collapse
|
27
|
Rihakova L, Quiniou C, Hamdan FF, Kaul R, Brault S, Hou X, Lahaie I, Sapieha P, Hamel D, Shao Z, Gobeil F, Hardy P, Joyal JS, Nedev H, Duhamel F, Beauregard K, Heveker N, Saragovi HU, Guillon G, Bouvier M, Lubell WD, Chemtob S. VRQ397 (CRAVKY): a novel noncompetitive V2 receptor antagonist. Am J Physiol Regul Integr Comp Physiol 2009; 297:R1009-18. [PMID: 19641130 DOI: 10.1152/ajpregu.90766.2008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Vasopressin type 2 receptor (V2R) exhibits mostly important properties for hydroosmotic equilibrium and, to a lesser extent, on vasomotricity. Drugs currently acting on this receptor are analogs of the natural neuropeptide, arginine vasopressin (AVP), and hence are competitive ligands. Peptides that reproduce specific sequences of a given receptor have lately been reported to interfere with its action, and if such molecules arise from regions remote from the binding site they would be anticipated to exhibit noncompetitive antagonism, but this has yet to be shown for V2R. Six peptides reproducing juxtamembranous regions of V2R were designed and screened; the most effective peptide, cravky (labeled VRQ397), was characterized. VRQ397 was potent (IC(50) = 0.69 +/- 0.25 nM) and fully effective in inhibiting V2R-dependent physiological function, specifically desmopressin-L-desamino-8-arginine-vasopressin (DDAVP)-induced cremasteric vasorelaxation; this physiological functional assay was utilized to avoid overlooking interference of specific signaling events. A dose-response profile revealed a noncompetitive property of VRQ397; correspondingly, VRQ397 bound specifically to V2R-expressing cells could not displace its natural ligand, AVP, but modulated AVP binding kinetics (dissociation rate). Specificity of VRQ397 was further confirmed by its inability to bind to homologous V1 and oxytocin receptors and its inefficacy to alter responses to stimulation of these receptors. VRQ397 exhibited pharmacological permissiveness on V2R-induced signals, as it inhibited DDAVP-induced PGI(2) generation but not that of cAMP or recruitment of beta-arrestin2. Consistent with in vitro and ex vivo effects as a V2R antagonist, VRQ397 displayed anticipated in vivo aquaretic efficacy. We hereby describe the discovery of a first potent noncompetitive antagonist of V2R, which exhibits functional selectivity, in line with properties of a negative allosteric modulator.
Collapse
Affiliation(s)
- L Rihakova
- Departments of Pediatrics and Pharmacology, Hôpital Ste Justine, Research Center, Montreal, QC, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Giniger RP, Buffat C, Millet V, Simeoni U. Renal effects of ibuprofen for the treatment of patent ductus arteriosus in premature infants. J Matern Fetal Neonatal Med 2009; 20:275-83. [PMID: 17437233 DOI: 10.1080/14767050701227950] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
In recent years ibuprofen has been proposed for the treatment of patent ductus arteriosus (PDA) as it has been proved to be equally as effective as indomethacin and shows fewer cerebral blood flow, intestinal and renal hemodynamic effects. A number of studies and several meta-analyses comparing both drugs are now available that debate whether indomethacin or ibuprofen should be used for PDA prophylaxis or closure. This review examines the available knowledge on the specific issue of the effects of ibuprofen on kidney function, as improved renal tolerance is a major argument in favor of its use in the routine treatment of PDA. There is sufficient evidence to consider that ibuprofen, at the currently proposed dosing regimen, has a similar efficacy to indomethacin but is better tolerated by the neonatal kidney when employed for the treatment of established PDA. However, adverse effects of ibuprofen have been evidenced both in trials on the use of ibuprofen for the prevention of PDA and of intraventricular hemorrhage-periventricular hemorrhage (IVH-PVH), and in experimental studies on a neonatal, anesthetized animal model. Thus ibuprofen, as with other cyclooxygenase (COX) inhibitors, may not be exempt from causing renal adverse effects, especially in circumstances when renal prostaglandin activation is maximal (i.e., when administrated early after birth, in more immature patients and in certain situations such as in the anesthetized rabbit). However, although the issue has been addressed extensively in the last decades, there is insufficient evidence that therapeutic intervention in PDA is beneficial in terms of mortality or clinically significant morbidity outcomes. Studies aimed at resolving this key issue are still needed.
Collapse
Affiliation(s)
- R P Giniger
- Faculté de Médecine, Université de la Méditerranée and Division of Neonatology La Conception Hospital, AP-HM, Marseille, France
| | | | | | | |
Collapse
|
29
|
Leblanc CP, Surette ME, Fiset S, Turgeon O'Brien H, Rioux FM. Maternal iron deficiency and its effect on essential fatty acid and eicosanoid metabolism and spatial memory in the guinea pig offspring. Prostaglandins Leukot Essent Fatty Acids 2009; 81:1-8. [PMID: 19524424 DOI: 10.1016/j.plefa.2009.05.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2008] [Revised: 04/25/2009] [Accepted: 05/21/2009] [Indexed: 10/20/2022]
Abstract
Iron deficiency is prevalent among infants and pregnant women in industrialized country. The goal of this study was to evaluate the impact of moderate maternal iron deficiency on the offspring's fatty acid and eicosanoid metabolism and spatial memory in guinea pigs. An iron-sufficient (IS) or iron-deficient (ID) diet was fed 14 days before mating and throughout pregnancy and lactation. The pups were tested for spatial memory on post-natal days 4-7. On post-natal day 9, the biochemical analysis included the pup's brain fatty acid profiles, prostaglandin (PGE(2) and PGF(2alpha)) concentrations and cyclooxygenase II protein levels. Spatial memory and indices of eicosanoid metabolism were comparable in both dietary groups. However, n-3 fatty acids were significantly higher (p<0.05) in brain of pups from the ID group. The data suggest that maternal iron deficiency results in a modification of the fatty acid profile of the offspring's brain that is not associated with any spatial memory deficits during early development.
Collapse
Affiliation(s)
- C P Leblanc
- Département des sciences des aliments et de nutrition, Faculté des sciences de l'agriculture et de l'alimentation, Université Laval, Québec, QC, Canada G1K 7P4
| | | | | | | | | |
Collapse
|
30
|
Wienecke T, Olesen J, Oturai PS, Ashina M. Prostaglandin E2 (PGE2) Induces Headache in Healthy Subjects. Cephalalgia 2009; 29:509-19. [DOI: 10.1111/j.1468-2982.2008.01748.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The role of prostanoids in nociception is well established. The headache-eliciting effects of prostaglandin E2 (PGE2) and its possible mechanisms have previously not been systematically studied in man. We hypothesized that infusion of PGE2 might induce headache and vasodilation of cranial vessels. PGE2 (0.40 μg kg−1 min−1) or saline was infused for 25 min into 11 healthy subjects in a cross-over, double-blind study. Headache intensity was scored on a verbal rating scale from 0 to 10. In addition, we recorded mean flow in the middle cerebral artery (VMCA) by transcranial Doppler and diameter of the superficial temporal artery (STA) by high-resolution ultrasonography. All 11 subjects reported headache on the PGE2 day and no subjects reported headache on the placebo day ( P = 0.001). During the immediate phase (0–30 min) ( P = 0.005) and the postinfusion phase (30–90 min) ( P = 0.005), the area under the curve for headache score was significantly larger on the PGE2 day compared with the placebo day. PGE2 caused dilatation of the STA (23.5%; 95% CI 14.0, 37.8) and the MCA (8.3%; 95% CI 4.0, 12.6). We suggest that PGE2 induces headache by activation and sensitization of cranial perivascular sensory afferents.
Collapse
Affiliation(s)
- T Wienecke
- Danish Headache Centre and Department of Neurology, University of Copenhagen, Glostrup, Copenhagen, Denmark
| | - J Olesen
- Danish Headache Centre and Department of Neurology, University of Copenhagen, Glostrup, Copenhagen, Denmark
| | - PS Oturai
- Department of Clinical Physiology and Nuclear Medicine, Glostrup Hospital, Faculty of Health Sciences, University of Copenhagen, Glostrup, Copenhagen, Denmark
| | - M Ashina
- Danish Headache Centre and Department of Neurology, University of Copenhagen, Glostrup, Copenhagen, Denmark
| |
Collapse
|
31
|
Andrade da Costa BLDS, Kang KD, Rittenhouse KD, Osborne NN. The localization of PGE2 receptor subtypes in rat retinal cultures and the neuroprotective effect of the EP2 agonist butaprost. Neurochem Int 2009; 55:199-207. [PMID: 19524109 DOI: 10.1016/j.neuint.2009.02.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Revised: 02/10/2009] [Accepted: 02/25/2009] [Indexed: 01/05/2023]
Abstract
It is concluded from immunohistochemical that all four types of prostaglandin-E(2) (PGE(2)) (EP1, EP2, EP3 and EP4) receptors are associated with specific cell-types in primary rat retinal cultures. Analysis specifically of EP2 receptor immunoreactivity shows it to coexist with some neurones expressing Thy-1 and calbindin immunoreactivities as well as with vimentin-positive Müller cells. Moreover, exposure of cultures to the EP2 specific agonist butaprost (100 nM) for a period of 24h results in a generation of cAMP thus providing support for the functionality of EP2 receptors. Cell survival was significantly affected in cultures where the serum concentration was reduced from 10 to 1% for 24h. This was reflected by a reduction in the number of GABA-positive neurons and an elevation of released lactate dehydrogenase (LDH) into the culture medium. Moreover, a number of cells displayed a clear generation of reactive oxygen species (ROS) and a staining for the breakdown of DNA by the TUNEL procedure as an indicator for apoptosis. These negative effects were attenuated when butaprost (100 nM) was present during the serum reduction and 30 min before the insult. The present studies provide evidence to show that all PGE(2) receptor types exist in the retina of rat pups, remain functional when the retinal cells are cultured and that specific activation of EP2 receptors with butaprost can attenuate a detrimental insult caused by insufficient serum that may occur in situ by reduced trophic support.
Collapse
Affiliation(s)
- Belmira Lara da Silveira Andrade da Costa
- Departamento de Fisiologia e Farmacologia, Centro de Ciências Biológicas (CCB), CCB-UFPE, Av. Prof. Moraes Rego s/n, Cidade Universitária, 50670901 Recife, PE, Brazil
| | | | | | | |
Collapse
|
32
|
|
33
|
Toscano CD, Kingsley PJ, Marnett LJ, Bosetti F. NMDA-induced seizure intensity is enhanced in COX-2 deficient mice. Neurotoxicology 2008; 29:1114-20. [PMID: 18834901 DOI: 10.1016/j.neuro.2008.08.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2008] [Accepted: 08/30/2008] [Indexed: 12/25/2022]
Abstract
Pharmacological inhibition or genetic deletion of cyclooxygenase (COX)-2, but not COX-1, has been shown to increase susceptibility to kainic acid (KA)-induced excitotoxicity. However, it is unclear if susceptibility to excitotoxins that act through other neurotransmitter receptors is altered by COX-2 inhibition. To further understand the involvement of COX-2 in regulating susceptibility to excitotoxicity, we investigated the effect of COX-2 deletion on excitotoxicity induced by peripheral injection of N-methyl-d-aspartate (NMDA, a specific agonist of the NMDA receptors) or lindane (a GABA(A) receptor antagonist). COX-2(-/-) mice injected intraperitoneally with NMDA (50-100mg/kg) exhibited significantly increased median seizure intensity when compared to COX-2(+/+) mice. Further, COX-2(-/-) mice exposed to NMDA showed neuronal damage, detected by Fluoro Jade B (FJB) staining, in the CA3 region of the hippocampus. There was no FJB staining nor any significant difference in median or maximal seizure intensity in COX-2(+/+) and COX-2(-/-) mice exposed to lindane. LC-MS/MS analysis of brain prostaglandin profile in COX-2(-/-) mice demonstrated a significant increase in PGF(2alpha), TXB(2), PGE(2) and PGD(2) expression 1h after administration of an excitotoxic dose of KA, but not of NMDA. Our findings demonstrate that COX-2 regulates susceptibility to KA and NMDA excitotoxicity, which directly activate glutamatergic neurotransmission, but not to lindane, which indirectly alters glutamatergic neurotransmission. Furthermore, increased levels of prostaglandins after seizures are associated with consistent manifestation of neuronal damage.
Collapse
Affiliation(s)
- Christopher D Toscano
- Molecular Neuroscience Unit, Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
34
|
Wei G, Kibler KK, Koehler RC, Maruyama T, Narumiya S, Doré S. Prostacyclin receptor deletion aggravates hippocampal neuronal loss after bilateral common carotid artery occlusion in mouse. Neuroscience 2008; 156:1111-7. [PMID: 18790018 DOI: 10.1016/j.neuroscience.2008.07.073] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2008] [Revised: 07/11/2008] [Accepted: 07/18/2008] [Indexed: 11/20/2022]
Abstract
Transient global cerebral ischemia causes delayed neuronal death in the hippocampal CA1 region. It also induces an increase in cyclooxygenase 2 (COX-2), which generates several metabolites of arachidonic acid, known as prostanoids, including prostacyclin (PGI(2)). To determine the role of the PGI(2) receptor (IP) in post-ischemic delayed cell death, wild-type and IP knockout (IP(-/-)) C57Bl/6 mice were subjected to 12-min bilateral common carotid artery occlusion or sham surgery, followed by 7 days of reperfusion. In the sham-operated mice, no statistical difference in CA1 hippocampal neuronal density was observed between the wild-type (2836+/-18/mm(2)) and IP(-/-) (2793+/-43/mm(2)) mice. Interestingly, in animals subjected to ischemia, surviving neuronal density in wild-type mice decreased to 50.5+/-7.9% and that of IP(-/-) mice decreased to 23.0+/-4.5% of their respective sham-operated controls (P<0.05). The results establish a role for the IP receptor in protecting pyramidal hippocampal neurons after this global ischemic model and suggest that IP receptor agonists could be developed to prevent delayed pyramidal neuronal cell death.
Collapse
Affiliation(s)
- G Wei
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | |
Collapse
|
35
|
Wang L, Pozzato V, Turato G, Madamanchi A, Murphy TM, Chitano P. Reduced spontaneous relaxation in immature guinea pig airway smooth muscle is associated with increased prostanoid release. Am J Physiol Lung Cell Mol Physiol 2008; 294:L964-73. [PMID: 18326825 DOI: 10.1152/ajplung.00401.2007] [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
Airway smooth muscle (ASM) from infant guinea pigs has less spontaneous relaxation during stimulation than ASM from adults. Inhibition of cyclooxygenase (COX), which catalyzes the production of prostanoids, increases this relaxation in infant ASM and abolishes age differences, thus suggesting that prostanoids reduce relaxation in infant ASM. In this study, we investigated whether leukotrienes are also involved in reducing spontaneous relaxation; whether the two COX isoforms, COX-1 and COX-2, differentially regulate spontaneous relaxation; and whether prostanoid release is developmentally regulated in guinea pig ASM. In different age groups, we measured relaxation during and after electrical stimulation in tracheal strips as well as prostanoid release from tracheal segments. Relaxation was studied in the absence and in the presence of a lipoxygenase inhibitor, a cysteinyl leukotriene receptor-1 antagonist, a COX-1 inhibitor, or a COX-2 inhibitor. We found that inhibition of lipoxygenase or cysteinyl leukotriene receptor-1 antagonism did not increase spontaneous relaxation at any age, thus excluding a role for leukotrienes in this phenomenon. Inhibition of COX-2, but not COX-1, promoted spontaneous relaxation. The basal release of prostanoids was more abundant in tissue from infant animals and decreased significantly with age. Thromboxane B2 was the most abundant metabolite released at all ages. Electrical stimulation and epithelium removal did not affect the age difference in prostanoid release. We conclude that increased basal prostanoid release contributes to the reduced spontaneous relaxation in immature guinea pig ASM compared with older animals. By regulating ASM relaxation, prostanoids may play a role in the airway hyperresponsiveness at a young age.
Collapse
Affiliation(s)
- Lu Wang
- Division of Pediatric Pulmonary Medicine, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | | | | | | | | | | |
Collapse
|
36
|
Kooli A, Kermorvant-Duchemin E, Sennlaub F, Bossolasco M, Hou X, Honoré JC, Dennery PA, Sapieha P, Varma D, Lachapelle P, Zhu T, Tremblay S, Hardy P, Jain K, Balazy M, Chemtob S. trans-Arachidonic acids induce a heme oxygenase-dependent vasorelaxation of cerebral microvasculature. Free Radic Biol Med 2008; 44:815-25. [PMID: 18082639 DOI: 10.1016/j.freeradbiomed.2007.11.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2007] [Revised: 10/24/2007] [Accepted: 11/07/2007] [Indexed: 10/22/2022]
Abstract
Nitrative stress is an important regulator of vascular tone. We have recently described that trans-arachidonic acids (TAA) are major products of NO(2)(.)-mediated isomerization of arachidonic acid in cell membranes and that nitrative stress increases TAA levels leading to neural microvascular degeneration. In the present study, we explored whether TAA exert acute effects on neuromicrovascular tone and investigated potential mechanisms thereof. TAA induced an endothelium-dependent vasorelaxation of rat brain pial microvasculature. This vasorelaxation was independent of nitric oxide, prostanoids, lipoxygenase products, and CYP(450) metabolite trans-hydroxyeicosatetraenoic acids. However, inhibition of heme oxygenase (using zinc protoporphyrin IX) and of dependent soluble guanylate cyclase (sGC; using ODQ) significantly diminished (by approximately 70%) the TAA-induced vasorelaxation. Consistent with these findings, TAA stimulated heme oxygenase (HO)-2-dependent bilirubin (using siRNA HO-2) and cGMP formation, and the HO product carbon monoxide (using CO-releasing CORM-2) reproduced the sGC-dependent cGMP formation and vasorelaxation. Further exploration revealed that TAA-induced vasorelaxation and bilirubin formation (HO activation) were nearly abrogated by large-conductance calcium-dependent potassium channels (BK(Ca)) (using TEA and iberiotoxin). Opening of BK(Ca) with the selective activator NS1619 induced a concentration-dependent vasorelaxation, which was inhibited by HO and sGC inhibitors. Coimmunoprecipitation suggested a molecular complex interaction between BK(Ca) and HO-2 (but not HO-1). Collectively, these findings identify new properties of TAA, specifically cerebral vasorelaxation through interactive activation of BK(Ca) with HO-2 and, in turn, sGC. Our findings provide new insights into the characterization of nitrative stress-derived TAA products, by showing they can act as acute mediators of nitrative stress on neurovascular tone.
Collapse
Affiliation(s)
- Amna Kooli
- Department of Paediatrics, Research Center of Hôpital Ste-Justine, Montréal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Non-selective cyclooxygenase inhibition before periodic acceleration (pGz) cardiopulmonary resuscitation (CPR) in a porcine model of ventricular fibrillation. Resuscitation 2008; 77:250-7. [PMID: 18241971 DOI: 10.1016/j.resuscitation.2007.11.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2007] [Revised: 10/30/2007] [Accepted: 11/12/2007] [Indexed: 01/13/2023]
Abstract
Whole body periodic acceleration (pGz) along the spinal axis is a novel method of cardiopulmonary resuscitation (CPR). Oscillatory motion of the supine body in a horizontal fashion provides ventilation and blood flow to vital organs during cardiac arrest and pulsatile shear stress to the vascular endothelium. We previously showed in pigs that pGz-CPR affords better overall survival, post resuscitation myocardial function, and neurological outcomes compared to conventional chest compression CPR. pGz through pulsatile shear stress on the vascular endothelium elicits acute production of prostaglandins and endothelial-derived nitric oxide (eNO) in whole animal models and in vitro preparations. The salutary effects associated with pGz-CPR compared to chest compression CPR are in part related to endothelial-derived nitric oxide. Both eNO and prostaglandins are cardioprotective in ischemia reperfusion models. To differentiate between the roles of these mediators, indomethacin a non-selective cyclooxygenase inhibitor (COX) was used as a tool to investigate prostaglandin effects during pGz-CPR by acute outcomes of survival, cardioprotection and regional blood flows (RBF). Two groups of anesthetized, intubated pigs weighing 25-36kg were studied. Prior to electrical induction of ventricular fibrillation (VF) animals received equal volumes of either saline placebo Control (CONT) (n=9) or indomethacin (INDO), (n=8), (2mg/kg). After 3min of unsupported VF, both groups received 15min of pGz-CPR followed by pharmacologic and electrical attempts for resuscitation. Return of circulation (ROSC) to 3h occurred in (78%) in CONT and (63%) in INDO pretreated animals. There was no statistically significant difference in hemodynamics between groups at baseline or during the protocol. At baseline, INDO caused a decrease in brain RBF. Two hours after ROSC, INDO blunted the hyperemia response to brain and heart. Echocardiographic evidence of myocardial dysfunction was most notable for the INDO group in the wall motion score index (WMSI). After 3h of ROSC there was a 4-fold difference in both creatine phosphokinase (CPK) and Troponin I concentration between INDO and CONT. Therefore, non-specific acute inhibition of COX in part blunts the salutary effects of pGz-CPR. These data suggest that prostaglandins in part are involved in the cardio protection induced by pGz during CPR.
Collapse
|
38
|
|
39
|
Role of Nitric Oxide and Prostaglandin Systems in Lithium Modulation of Acetylcholine Vasodilation. J Cardiovasc Pharmacol 2007; 50:641-6. [DOI: 10.1097/fjc.0b013e318153f262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
40
|
Finkel JC. Use of nonsteroidal anti inflammatory drugs in preterm, term neonates and infants: analgesia by consensus? Paediatr Anaesth 2007; 17:915-7. [PMID: 17767625 DOI: 10.1111/j.1460-9592.2007.02320.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
41
|
Afsharimani B, Moezi L, Sadeghipour H, Rahimzadeh-Rofouyi B, Nobakht M, Sanatkar M, Ghahremani MH, Dehpour AR. Effect of chronic lithium administration on endothelium-dependent relaxation of rat mesenteric bed: role of nitric oxide. Can J Physiol Pharmacol 2007; 85:1038-46. [DOI: 10.1139/y07-095] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The mechanism of action of lithium, an effective treatment for bipolar disease, is still unknown. In this study, the mesenteric vascular beds of control rats and rats that were chronically treated with lithium were prepared by the McGregor method, and the mesenteric vascular bed vasorelaxation responses were examined. NADPH-diaphorase histochemistry was used to determine the activity of NOS (nitric oxide synthase) in mesenteric vascular beds. We demonstrated that ACh-induced vasorelaxation increased in the mesenteric vascular bed of rats treated with lithium. Acute Nο-nitro-l-arginine methyl ester (l-NAME) administration in the medium blocked ACh-induced vasorelaxation in the control group more effectively than in lithium-treated rats, while the vasorelaxant response to sodium nitroprusside, a NO donor, was not different between lithium-treated and control groups. Acute aminoguanidine administration blocked ACh-induced vasorelaxation of lithium-treated rats, but had no effect in the control rats. Furthermore, NOS activity, determined by NADPH-diaphorase staining, was significantly greater in the mesenteric vascular beds from chronic lithium-treated rats than in those from control rats. These data suggest that the enhanced ACh-induced endothelium-derived vasorelaxation in rat mesenteric bed from chronic lithium-treated rats might be associated with increased NOS activity, likely via iNOS. Simultaneous acute l-NAME and indomethacin administration suggests the possible upregulation of EDHF (endothelium-derived hyperpolarizing factor) in lithium-treated rats.
Collapse
Affiliation(s)
- Banafsheh Afsharimani
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
- Department of Pharmacology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Anatomy and Histology Department, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Basic Sciences Research Centre, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Moezi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
- Department of Pharmacology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Anatomy and Histology Department, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Basic Sciences Research Centre, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Sadeghipour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
- Department of Pharmacology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Anatomy and Histology Department, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Basic Sciences Research Centre, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Rahimzadeh-Rofouyi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
- Department of Pharmacology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Anatomy and Histology Department, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Basic Sciences Research Centre, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Maliheh Nobakht
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
- Department of Pharmacology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Anatomy and Histology Department, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Basic Sciences Research Centre, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Sanatkar
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
- Department of Pharmacology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Anatomy and Histology Department, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Basic Sciences Research Centre, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hosein Ghahremani
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
- Department of Pharmacology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Anatomy and Histology Department, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Basic Sciences Research Centre, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad R. Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
- Department of Pharmacology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Anatomy and Histology Department, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Basic Sciences Research Centre, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
42
|
Antonucci R, Cuzzolin L, Arceri A, Fanos V. Urinary prostaglandin E2 in the newborn and infant. Prostaglandins Other Lipid Mediat 2007; 84:1-13. [PMID: 17643883 DOI: 10.1016/j.prostaglandins.2007.04.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2006] [Revised: 04/22/2007] [Accepted: 04/23/2007] [Indexed: 10/23/2022]
Abstract
Prostaglandin E(2) (PGE(2)) belongs to a family of biologically active lipids derived from the 20-carbon essential fatty acids. Renal PGE(2) is involved in the development of the kidney; it also contributes to regulate renal perfusion and glomerular filtration rate, and controls water and electrolyte balance. Furthermore, this mediator protects the kidney against excessive functional changes during the transition from fetal to extrauterine life, when it counteracts the vasoconstrictive effects of high levels of angiotensin II and other mediators. There is evidence that PGE(2) plays an important pathophysiological role in neonatal conditions of renal stress, and in congenital or acquired nephropaties. Thus, measurement of urinary PGE(2) as an index of renal synthesis of this primary prostaglandin may represent a non-invasive and sensitive method of investigating the homeostatic function of the kidney in early life. The aim of this literature review is to examine urinary PGE(2) as a non-invasive marker of renal homeostasis in the newborn and infant under both physiological and pathological conditions, or during treatments with widely used, potentially toxic drugs.
Collapse
Affiliation(s)
- Roberto Antonucci
- Department of Pediatrics and Clinical Medicine-Section of Neonatal Intensive Care Unit, University of Cagliari, Cagliari, Italy
| | | | | | | |
Collapse
|
43
|
Lugo B, Ford HR, Grishin A. Molecular signaling in necrotizing enterocolitis: regulation of intestinal COX-2 expression. J Pediatr Surg 2007; 42:1165-71. [PMID: 17618875 DOI: 10.1016/j.jpedsurg.2007.02.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Necrotizing enterocolitis (NEC) is the most common surgical emergency in premature infants. The underlying etiology of NEC remains unknown, although bacterial colonization of the gut, formula feeding, and perinatal stress have been implicated as putative risk factors. The disease is characterized by exuberant gut inflammation leading to ischemia and coagulation necrosis of the intestinal epithelium. The molecular and cellular mechanisms responsible for these pathologic changes are poorly understood. It has been shown that various exogenous and endogenous mediators such as lipopolysaccharide, inflammatory cytokines, platelet activating factor, and nitric oxide may play a role in the pathogenesis of NEC. Recent studies in our laboratory and others have established a link between NEC and activation of cyclooxygenase-2, the enzyme that catalyzes the rate-limiting step in the biosynthesis of prostanoids. The challenge is in defining the molecular signaling pathways leading to accumulation of these mediators early in the disease progression, before the onset of tissue necrosis and systemic sepsis. Identification and characterization of these pathways could lead to the development of novel treatment strategies to alleviate the morbidity and mortality associated with NEC.
Collapse
Affiliation(s)
- Brian Lugo
- Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | | | | |
Collapse
|
44
|
Doheny HC, O'Reilly MJ, Sexton DJ, Morrison JJ. THG113.31, a specific PGF2alpha receptor antagonist, induces human myometrial relaxation and BKCa channel activation. Reprod Biol Endocrinol 2007; 5:10. [PMID: 17367527 PMCID: PMC1831777 DOI: 10.1186/1477-7827-5-10] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2007] [Accepted: 03/16/2007] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND PGF2alpha exerts a significant contractile effect on myometrium and is central to human labour. THG113.31, a specific non-competitive PGF2alpha receptor (FP) antagonist, exerts an inhibitory effect on myometrial contractility. The BKCa channel is ubiquitously encountered in human uterine tissue and plays a significant role in modulating myometrial cell membrane potential and excitability. The objective of this study was to investigate potential BKCa channel involvement in the response of human myometrium to THG113.31. METHODS Single and whole-cell electrophysiological BKCa channel recordings from freshly dispersed myocytes, were investigated in the presence and absence of THG113.31. Functional studies investigated the effects of THG113.31 on isolated spontaneous myometrial contractions, in the presence and absence of the BKCa channel blocker, iberiotoxin. RESULTS Single channel recordings identified the BKCa channel as a target of THG113.31. THG113.31 significantly increased the open state probability of these channels [control 0.023+/-0.006; 10 microM THG113.31 0.087+/-0.012 (P = 0.009); and 50 microM THG113.31 0.1356+/-0.018 (P = 0.001)]. In addition, THG113.31 increased whole-cell BKCa currents over a range of membrane potentials, and this effect was reversed by 100 nanoM IbTX. Isometric tension studies demonstrated that THG113.31 exerted a significant concentration-dependent relaxant effect on human myometrial tissue and pre-incubation of strips with IbTX abolished this effect on spontaneously occurring contractions. CONCLUSION These data suggests that activation of the BKCa channel may contribute, at least partially, to the uterorelaxant effect of THG113.31.
Collapse
Affiliation(s)
- Helen C Doheny
- Department of Obstetrics & Gynaecology, National University of Ireland Galway, Clinical Science Institute, University College Hospital Galway, Newcastle Road, Galway, Ireland
| | - Michael J O'Reilly
- Department of Obstetrics & Gynaecology, National University of Ireland Galway, Clinical Science Institute, University College Hospital Galway, Newcastle Road, Galway, Ireland
| | - Donal J Sexton
- Department of Obstetrics & Gynaecology, National University of Ireland Galway, Clinical Science Institute, University College Hospital Galway, Newcastle Road, Galway, Ireland
| | - John J Morrison
- Department of Obstetrics & Gynaecology, National University of Ireland Galway, Clinical Science Institute, University College Hospital Galway, Newcastle Road, Galway, Ireland
| |
Collapse
|
45
|
Nishida N, Blood AB, Hunter CJ, Bragg S, Williams J, Pearce WJ, Power GG. Role of prostanoids in the regulation of cerebral blood flow during normoxia and hypoxia in the fetal sheep. Pediatr Res 2006; 60:524-9. [PMID: 16988195 DOI: 10.1203/01.pdr.0000242268.99726.53] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The fetal cardiovascular responses to hypoxia include decreased peripheral blood flow and increased cerebral, cardiac, and adrenal blood flow. Prostanoids, metabolites of cyclooxygenase enzyme activity, have potent effects on vascular tone in both the adult and the fetus. To examine the role of prostanoids in the regulation of fetal cerebral blood flow (CBF) during acute hypoxic stress, eight near term fetal sheep were studied after infusing vehicle or diclofenac, a cyclooxygenase inhibitor, followed by a 30-min period of hypoxia (arterial Po(2) 12 Torr). In the control experiments, CBF, measured continuously with laser Doppler flowmetry, increased to 148% of baseline values (p < 0.01) and cerebral vascular resistance decreased to 70% of baseline values after 30 min of hypoxic stress. During diclofenac infusion, hypoxia resulted in a CBF increase to only 129% of baseline, a significant attenuation (p < 0.05), accompanied by decreased plasma prostanoid concentrations. Increases in mean arterial blood pressure during hypoxia were also attenuated by diclofenac infusion. Flow and pressure responses were not accompanied by changes in cerebral vascular resistance. These results indicate that prostanoids indirectly modulate fetal CBF responses to hypoxia, but that their effects are mediated through modulation of systemic rather than cerebral vascular tone.
Collapse
Affiliation(s)
- Naoko Nishida
- Department of Obstetrics and Gynecology, Nippon Medical School, Tokyo, Japan 13-8602
| | | | | | | | | | | | | |
Collapse
|
46
|
Schmitz T, Levine BA, Nathanielsz PW. Localization and steroid regulation of prostaglandin E2 receptor protein expression in ovine cervix. Reproduction 2006; 131:743-50. [PMID: 16595725 DOI: 10.1530/rep.1.00767] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Although prostaglandin E2 (PGE2) has been identified as a central mediator of the cervical ripening process, the mechanisms responsible for PGE2 ripening are still poorly understood, partly because of the lack of information concerning the precise cellular localization and regulation of PGE2 (EP) receptors in the cervix. To provide new insights into the mechanisms of cervical ripening, we used indirect immunofluorescence to localize cervical EP receptor protein expression in ovariectomized ewes and examined the effect of administration of progesterone or estradiol. EP receptors were widely distributed in cervical blood vessels, epithelium of the cervical canal, circular and longitudinal muscles, and stroma. Estradiol replacement decreased EP1 and EP3 receptor protein in blood vessel media (by 23 and 31% respectively, P < 0.05) and decreased EP1 receptor protein expression in the longitudinal muscle layer (by 27%, P < 0.05). Stromal EP1 and EP3 receptor protein expression was also reduced by estradiol (by 29 and 20% respectively, P < 0.05). Progesterone replacement had no significant effect on EP receptor protein expression. The arterial changes would favor PGE2-induced vasodilatation, subsequent edema and leukocyte infiltration during the cervical ripening process whereas the muscular alterations would facilitate smooth muscle relaxation and cervical dilatation. Furthermore, estradiol provoked perinuclear localization of EP3 receptor protein in the longitudinal muscle layer. This latter result suggests that cellular EP receptor localization is regulated by estradiol and that PGE2 may also control smooth muscle contraction and regulate ovine cervical dilatation in an intracrine manner via EP3 receptors.
Collapse
Affiliation(s)
- Thomas Schmitz
- Maternité Port-Royal, Hopital Cochin, AP-HP, Université René Descartes Paris V, Boulevard de Port-Royal, 75014 Paris, France.
| | | | | |
Collapse
|
47
|
Phillis JW, Horrocks LA, Farooqui AA. Cyclooxygenases, lipoxygenases, and epoxygenases in CNS: Their role and involvement in neurological disorders. ACTA ACUST UNITED AC 2006; 52:201-43. [PMID: 16647138 DOI: 10.1016/j.brainresrev.2006.02.002] [Citation(s) in RCA: 266] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2005] [Revised: 02/23/2006] [Accepted: 02/24/2006] [Indexed: 01/01/2023]
Abstract
Three enzyme systems, cyclooxygenases that generate prostaglandins, lipoxygenases that form hydroxy derivatives and leukotrienes, and epoxygenases that give rise to epoxyeicosatrienoic products, metabolize arachidonic acid after its release from neural membrane phospholipids by the action of phospholipase A(2). Lysophospholipids, the other products of phospholipase A(2) reactions, are either reacylated or metabolized to platelet-activating factor. Under normal conditions, these metabolites play important roles in synaptic function, cerebral blood flow regulation, apoptosis, angiogenesis, and gene expression. Increased activities of cyclooxygenases, lipoxygenases, and epoxygenases under pathological situations such as ischemia, epilepsy, Alzheimer's disease, Parkinson disease, amyotrophic lateral sclerosis, and Creutzfeldt-Jakob disease produce neuroinflammation involving vasodilation and vasoconstriction, platelet aggregation, leukocyte chemotaxis and release of cytokines, and oxidative stress. These are closely associated with the neural cell injury which occurs in these neurological conditions. The metabolic products of docosahexaenoic acid, through these enzymes, generate a new class of lipid mediators, namely docosatrienes and resolvins. These metabolites antagonize the effect of metabolites derived from arachidonic acid. Recent studies provide insight into how these arachidonic acid metabolites interact with each other and other bioactive mediators such as platelet-activating factor, endocannabinoids, and docosatrienes under normal and pathological conditions. Here, we review present knowledge of the functions of cyclooxygenases, lipoxygenases, and epoxygenases in brain and their association with neurodegenerative diseases.
Collapse
Affiliation(s)
- John W Phillis
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA.
| | | | | |
Collapse
|
48
|
Rheinlaender C, Weber SCT, Sarioglu N, Strauss E, Obladen M, Koehne P. Changing expression of cyclooxygenases and prostaglandin receptor EP4 during development of the human ductus arteriosus. Pediatr Res 2006; 60:270-5. [PMID: 16857763 DOI: 10.1203/01.pdr.0000233066.28496.7c] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Programmed proliferative degeneration of the human fetal ductus arteriosus (DA) in preparation for its definite postnatal closure has a large developmental variability and is controlled by several signaling pathways, most prominently by prostaglandin (PG) metabolism. Numerous studies in various mammalian species have shown interspecies and developmental differences in ductal protein expression of cyclooxygenase (COX) isoforms and PG E receptor subtypes (EP1-4). We examined COX1, COX2, and EP4 receptor protein expression immunohistochemically in 57 human fetal autopsy DA specimens of 11-38 wk of gestation. According to their histologic maturity, specimens were classified into four stages using a newly designed maturity score that showed that histologic maturity of the DA was not closely related to gestational age. COX1 expression was found in all DA regions and rose steadily during development. COX2 staining remained weak throughout gestation. EP4 receptor staining increased moderately during gestation and was limited to the intima and media. In conclusion, histologic maturity classification helps to address developmentally regulated processes in the fetal DA. Concerning prostaglandin metabolism our findings are in line with animal studies, which assigned COX1 the predominant role in the DA throughout gestation. EP4 receptor presumably plays a key role for active patency of the human DA in the third trimester.
Collapse
Affiliation(s)
- Cornelia Rheinlaender
- Department of Neonatology, Charité Universitätsmedizin Berlin, Campus Virchow Hospital, 13353 Berlin, Germany
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
Ibuprofen, a nonsteroidal antiinflammatory drug, widely used as antipyretic, antiinflammatory, and analgesic agent and for therapy of arthritis, exerts a dose-dependent constriction of the ductus arteriosus in newborn lambs. Two intravenous preparations, namely ibuprofen lysine and ibuprofen-THAM, have been studied in preterm newborns with patent ductus arteriosus. Clinical trials have compared IV ibuprofen to placebo, or to indomethacin. Pharmacodynamic effects of this drug before and after its administration have also been evaluated. Compared with placebo, IV ibuprofen effectively closed PDA with minimal effect on renal function. One study using intravenous ibuprofen-THAM showed decreased renal function and increased risk of NEC and PPHN. Compared with indomethacin, IV ibuprofen lysine exerted similar efficacy (75% to 93% closure). However, indomethacin increased abnormal renal function and decreased mesenteric and cerebral blood flow and bio-energetics. Two clinical trials showed that ibuprofen did not reduce the incidence of intraventricular hemorrhage compared with placebo. The drug has prolonged elimination (plasma half-life = ca 23 hours), suggesting that once daily dosing is appropriate. Dose finding studies indicate that a starting dose of 10 mg/kg followed by 5 mg/kg/d for 2 more days provides optimal efficacy with the least adverse effects. Neonatal data on ibuprofen and indomethacin indicate that, on the first day of life when IVH prevention is desired, indomethacin and not ibuprofen should be used since ibuprofen has no effect on IVH risk. On or after the second day of postnatal life, when early or therapeutic PDA closure is needed, ibuprofen and not indomethacin is probably the first choice due to its better adverse event profile.
Collapse
Affiliation(s)
- J V Aranda
- Pediatric Pharmacology Research Unit Network (PPRU), Division of Clinical Pharmacology and Toxicology, Children's Hospital of Michigan, 3901 Beaubien Boulevard 3N47, Detroit, MI 48201, USA.
| | | |
Collapse
|
50
|
Krotz F, Schiele TM, Klauss V, Sohn HY. Selective COX-2 inhibitors and risk of myocardial infarction. J Vasc Res 2005; 42:312-24. [PMID: 15976506 DOI: 10.1159/000086459] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2005] [Accepted: 04/08/2005] [Indexed: 12/31/2022] Open
Abstract
Selective inhibitors of cyclooxygenase-2 (COX-2, 'coxibs') are highly effective anti-inflammatory and analgesic drugs that exert their action by preventing the formation of prostanoids. Recently some coxibs, which were designed to exploit the advantageous effects of non-steroidal anti-inflammatory drugs while evading their side effects, have been reported to increase the risk of myo cardial infarction and atherothrombotic events. This has led to the withdrawal of rofecoxib from global markets, and warnings have been issued by drug authorities about similar events during the use of celecoxib or valdecoxib/parecoxib, bringing about questions of an inherent atherothrombotic risk of all coxibs and consequences that should be drawn by health care professionals. These questions need to be addressed in light of the known effects of selective inhibition of COX-2 on the cardiovascular system. Although COX-2, in contrast to the cyclooxygenase-1 (COX-1) isoform, is regarded as an inducible enzyme that only has a role in pathophysiological processes like pain and inflammation, experimental and clinical studies have shown that COX-2 is constitutively expressed in tissues like the kidney or vascular endothelium, where it executes important physiological functions. COX-2-dependent formation of prostanoids not only results in the mediation of pain or inflammatory signals but also in the maintenance of vascular integrity. Especially prostacyclin (PGI(2)), which exerts vasodilatory and antiplatelet properties, is formed to a significant extent by COX-2, and its levels are reduced to less than half of normal when COX-2 is inhibited. This review outlines the rationale for the development of selective COX-2 inhibitors and the pathophysiological consequences of selective inhibition of COX-2 with special regard to vasoactive prostaglandins. It describes coxibs that are current ly available, evaluates the current knowledge on the risk of atherothrombotic events associated with their intake and critically discusses the consequences that should be drawn from these insights.
Collapse
Affiliation(s)
- Florian Krotz
- Institute of Cardiology, Medical Polyclinic, Ludwig Maximilians University, Munich, Germany.
| | | | | | | |
Collapse
|