Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Fr Ophtalmol ; 38(10): 983-95, 2015 Dec.
Article in French | MEDLINE | ID: mdl-26597554

ABSTRACT

The exact pathophysiology of glaucoma is not fully understood. Understanding of the vascular pathophysiology of glaucoma requires: knowing the techniques for measuring ocular blood flow and characterizing the topography of vascular disease and the mechanisms involved in this neuropathy. A decreased mean ocular perfusion pressure and a loss of vascular autoregulation are implicated in glaucomatous disease. Early decrease in ocular blood flow has been identified in primary open-angle glaucoma and normal pressure glaucoma, contributing to the progression of optic neuropathy. The vascular damage associated with glaucoma is present in various vascular territories within the eye (from the ophthalmic artery to the retina) and is characterized by a decrease in basal blood flow associated with a dysfunction of vasoregulation.


Subject(s)
Glaucoma/physiopathology , Hemodynamics , Angiotensin II/physiology , Arterial Pressure , Blood Viscosity , Endothelin-1/physiology , Endothelium, Vascular/physiopathology , Eye/blood supply , Humans , Intraocular Pressure , Nitric Oxide/physiology , Prostaglandins I/physiology , Vascular Resistance , Vasoconstriction/physiology , Vasodilation/physiology , Vasomotor System/physiopathology
2.
Endocrinology ; 156(2): 499-510, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25406016

ABSTRACT

Prostaglandins, particularly PGE2, are important to adult bone and joint health, but how prostaglandins act on growth plate cartilage to affect bone growth is unclear. We show that growth plate cartilage is distinct from articular cartilage with respect to cyclooxygenase (COX)-2 mRNA expression; although articular chondrocytes express very little COX-2, COX-2 expression is high in growth plate chondrocytes and is increased by IGF-I. In bovine primary growth plate chondrocytes, ATDC5 cells, and human metatarsal explants, inhibition of COX activity with nonsteroidal antiinflammatory drugs (NSAIDs) inhibits chondrocyte proliferation and ERK activation by IGF-I. This inhibition is reversed by prostaglandin E2 and prostacyclin (PGI2) but not by prostaglandin D2 or thromboxane B2. Inhibition of COX activity in young mice by ip injections of NSAIDs causes dwarfism. In growth plate chondrocytes, inhibition of proliferation and ERK activation by NSAIDs is reversed by forskolin, 8-bromoadenosine, 3',5'-cAMP and a prostacyclin analog, iloprost. The inhibition of proliferation and ERK activation by celecoxib is also reversed by 8CPT-2Me-cAMP, an activator of Epac, implicating the small G protein Rap1 in the pathway activated by iloprost. These results imply that prostacyclin is required for proper growth plate development and bone growth.


Subject(s)
Bone Development , Guanine Nucleotide Exchange Factors/metabolism , Prostaglandins I/physiology , rap1 GTP-Binding Proteins/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal , Cattle , Celecoxib , Cells, Cultured , Chondrocytes/enzymology , Cyclooxygenase 2/metabolism , Epoprostenol/metabolism , Female , Growth Plate/drug effects , Growth Plate/enzymology , Humans , Iloprost , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Inbred C57BL , Pyrazoles , Signal Transduction , Sulfonamides
3.
J Gastroenterol Hepatol ; 29 Suppl 4: 3-10, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25521725

ABSTRACT

BACKGROUND AND AIM: We investigated the roles of cyclooxygenase (COX) isozymes and prostaglandins (PGs) and their receptors in mucosal defense against cold-restraint stress (CRS)-induced gastric lesions. METHODS: Male C57BL/6 wild-type (WT) mice and those lacking COX-1 or COX-2 as well as those lacking EP1, EP3, or IP receptors were used after 18 h fasting. Animals were restrained in Bollman cages and kept in a cold room at 10°C for 90 min. RESULTS: CRS induced multiple hemorrhagic lesions in WT mouse stomachs. The severity of these lesions was significantly worsened by pretreatment with the nonselective COX inhibitors (indomethacin, loxoprofen) or selective COX-1 inhibitor (SC-560), while neither of the selective COX-2 inhibitors (rofecoxib and celecoxib) had any effect. These lesions were also aggravated in animals lacking COX-1, but not COX-2. The expression of COX-2 mRNA was not detected in the stomach after CRS, while COX-1 expression was observed under normal and stressed conditions. The gastric ulcerogenic response to CRS was similar between EP1 or EP3 knockout mice and WT mice, but was markedly worsened in animals lacking IP receptors. Pretreating WT mice with iloprost (the PGI2 analog) significantly prevented CRS-induced gastric lesions in the presence of indomethacin. PGE2 also reduced the severity of these lesions, and the effect was mimicked by the EP4 agonist, AE1-329. CONCLUSIONS: These results suggest that endogenous PGs derived from COX-1 play a crucial role in gastric mucosal defense during CRS, and this action is mainly mediated by PGI2 /IP receptors and partly by PGE2 /EP4 receptors.


Subject(s)
Cold Temperature/adverse effects , Cyclooxygenase 1/physiology , Cyclooxygenase Inhibitors/adverse effects , Gastric Mucosa/pathology , Gastrointestinal Hemorrhage/etiology , Gastrointestinal Hemorrhage/pathology , Prostaglandins I/physiology , Stress, Physiological/physiology , Animals , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Dinoprostone/physiology , Epoprostenol/physiology , Gene Expression , Indomethacin/adverse effects , Male , Mice, Inbred C57BL , Phenylpropionates/adverse effects , Pyrazoles/adverse effects , RNA, Messenger/metabolism , Receptors, Epoprostenol/physiology , Receptors, Prostaglandin E/physiology , Receptors, Prostaglandin E, EP4 Subtype/physiology
4.
Anesthesiology ; 120(2): 447-58, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23969560

ABSTRACT

BACKGROUND: Prostacyclin (PGI2) is known to be an important mediator of peripheral pain sensation (nociception) whereas little is known about its role in central sensitization. METHODS: The levels of the stable PGI2-metabolite 6-keto-prostaglandin F1α (6-keto-PGF1α) and of prostaglandin E2 (PGE2) were measured in the dorsal horn with the use of mass spectrometry after peripheral inflammation. Expression of the prostanoid receptors was determined by immunohistology. Effects of prostacyclin receptor (IP) activation on spinal neurons were investigated with biochemical assays (cyclic adenosine monophosphate-, glutamate release-measurement, Western blot analysis) in embryonic cultures and adult spinal cord. The specific IP antagonist Cay10441 was applied intrathecally after zymosan-induced mechanical hyperalgesia in vivo. RESULTS: Peripheral inflammation caused a significant increase of the stable PGI2 metabolite 6-keto-PGF1α in the dorsal horn of wild-type mice (n = 5). IP was located on spinal neurons and did not colocalize with the prostaglandin E2 receptors EP2 or EP4. The selective IP-agonist cicaprost increased cyclic adenosine monophosphate synthesis in spinal cultures from wild-type but not from IP-deficient mice (n = 5-10). The combination of fluorescence-resonance-energy transfer-based cyclic adenosine monophosphate imaging and calcium imaging showed a cicaprost-induced cyclic adenosine monophosphate synthesis in spinal cord neurons (n = 5-6). Fittingly, IP activation increased glutamate release from acute spinal cord sections of adult mice (n = 13-58). Cicaprost, but not agonists for EP2 and EP4, induced protein kinase A-dependent phosphorylation of the GluR1 subunit and its translocation to the membrane. Accordingly, intrathecal administration of the IP receptor antagonist Cay10441 had an antinociceptive effect (n = 8-11). CONCLUSION: Spinal prostacyclin synthesis during early inflammation causes the recruitment of GluR1 receptors to membrane fractions, thereby augmenting the onset of central sensitization.


Subject(s)
Cyclic AMP/physiology , Nociception/physiology , Prostaglandins I/physiology , Receptors, AMPA/metabolism , Spinal Cord/physiology , Animals , Behavior, Animal/drug effects , Blotting, Western , Calcium/metabolism , Chromatography, High Pressure Liquid , Epitopes , Female , Fluorescence Resonance Energy Transfer , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Neurons/physiology , Pain/psychology , Pregnancy , Prostaglandins I/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Spinal Cord/cytology , Spinal Cord/metabolism , Tandem Mass Spectrometry , Translocation, Genetic
6.
Circulation ; 126(9): 1067-78, 2012 Aug 28.
Article in English | MEDLINE | ID: mdl-22855570

ABSTRACT

BACKGROUND: Perivascular adipose tissue (PVAT) surrounds most vessels and shares common features with brown adipose tissue (BAT). Although adaptive thermogenesis in BAT increases energy expenditure and is beneficial for metabolic diseases, little is known about the role of PVAT in vascular diseases such as atherosclerosis. We hypothesize that the thermogenic function of PVAT regulates intravascular temperature and reduces atherosclerosis. METHODS AND RESULTS: PVAT shares similar structural and proteomics with BAT. We demonstrated that PVAT has thermogenic properties similar to BAT in response to cold stimuli in vivo. Proteomics analysis of the PVAT from mice housed in a cold environment identified differential expression in proteins highly related to cellular metabolic processes. In a mouse model deficient in peroxisome proliferator-activated receptor-γ in smooth muscle cells (SMPG KO mice), we uncovered a complete absence of PVAT surrounding the vasculature, likely caused by peroxisome proliferator-activated receptor-γ deletion in the perivascular adipocyte precursor cells as well. Lack of PVAT, which results in loss of its thermogenic activity, impaired vascular homeostasis, which caused temperature loss and endothelial dysfunction. We further showed that cold exposure inhibits atherosclerosis and improves endothelial function in mice with intact PVAT but not in SMPG KO mice as a result of impaired lipid clearance. Proinflammatory cytokine expression in PVAT is not altered on exposure to cold. Finally, prostacyclin released from PVAT contributes to the vascular protection against endothelial dysfunction. CONCLUSIONS: PVAT is a vasoactive organ with functional characteristics similar to BAT and is essential for intravascular thermoregulation of cold acclimation. This thermogenic capacity of PVAT plays an important protective role in the pathogenesis of atherosclerosis.


Subject(s)
Adipose Tissue/physiopathology , Atherosclerosis/etiology , Body Temperature Regulation/physiology , Muscle, Smooth, Vascular/physiopathology , PPAR gamma/deficiency , Adaptation, Physiological/physiology , Adipocytes/metabolism , Adipose Tissue/pathology , Adipose Tissue, Brown/metabolism , Animals , Aorta , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/prevention & control , Carotid Arteries , Cold Temperature , Cytokines/metabolism , Diet, Atherogenic , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Gene Expression Regulation/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , PPAR gamma/genetics , Prostaglandins I/physiology , Proteomics
7.
Rev Med Suisse ; 7(321): 2435-8, 2011 Dec 14.
Article in French | MEDLINE | ID: mdl-22279861

ABSTRACT

The hemodynamics of septic shock is characterized by a primary reduction of vascular tone, which defines vasoplegia. Septic vasoplegia is due to reduced endogenous production of vasopressin, as well as to the overproduction of vasodilating molecules (nitric oxide, prostacyclin, peroxynitrite and kynurenine) and the opening of ATP-sensitive potassium channels. Treatment is supportive and includes primarily alpha-adrenergic catecholamines. Vasopressin may also be useful, although its place is still controversial. Further agents can improve the vascular responsiveness to catecholamines, most notably low doses hydrocortisone, and, to a lesser extent, activated protein C. Further, innovative therapies, based on recent understanding of pathophysiological mechanisms, might become useful agents to treat septic vasoplegia in the future.


Subject(s)
Shock, Septic/complications , Shock, Septic/therapy , Vasoplegia/etiology , Vasoplegia/therapy , Catecholamines/therapeutic use , Humans , Hydrocortisone/therapeutic use , KATP Channels/metabolism , KATP Channels/physiology , Models, Biological , Nitric Oxide/adverse effects , Nitric Oxide/metabolism , Nitric Oxide/physiology , Peroxynitrous Acid/adverse effects , Peroxynitrous Acid/metabolism , Peroxynitrous Acid/physiology , Prostaglandins I/adverse effects , Prostaglandins I/metabolism , Prostaglandins I/physiology , Protein C/therapeutic use , Shock, Septic/metabolism , Signal Transduction/physiology , Vasoplegia/metabolism , Vasopressins/therapeutic use
9.
Br J Pharmacol ; 146(7): 983-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16231006

ABSTRACT

The object of the present study was to clarify the neurotransmitter(s) controlling membrane responses to electrical field stimulation (EFS) in the circular smooth muscle cells of first-order branches of chicken anterior mesenteric artery.EFS (five pulses at 20 Hz, 1 ms) evoked a hyperpolarization of amplitude--21.6+/-1.2 mV, total duration 21.8+/-1.2 s and latency 641.7+/-81.9 ms. The response was tetrodotoxin-sensitive and nonadrenergic noncholinergic (NANC) in nature. The NANC response was blocked by the nonspecific purinergic antagonist, suramin, indicating that the response is mediated by the neurotransmitter adenosine 5'-triphosphate (ATP). Either desensitization or blockade of P2Y receptor with its putative agonist 2-methylthioATP (1 microM for 30 min) or with its antagonist cibacron blue F3GA (10 microM), respectively, abolished the purinergic hyperpolarization. PPADS at concentrations up to 100 microM had no effect on the EFS-induced response, indicating that this response is mediated through P2Y, but not P2X, receptor. In addition, the response was completely abolished by two specific P2Y1 receptor antagonists, namely, MRS 2179 (300 nM) and A3P5PS (10 microM). Removal of the endothelium abolished the purinergic hyperpolarization, which was converted, in some preparations, to a small depolarization, indicating that the hyperpolarizing response is endothelium-dependent. The present study suggests that in first-order branches of chicken anterior mesenteric artery, ATP released from perivascular nerves may diffuse to the endothelium-activating P2Y1 receptor to induce release of an inhibitory substance that mediates hyperpolarization in the circular smooth muscle.


Subject(s)
Adenosine Triphosphate/physiology , Endothelium, Vascular/physiology , Mesenteric Arteries/physiology , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Animals , Chickens , Electric Stimulation , Female , In Vitro Techniques , Indomethacin/pharmacology , Membrane Potentials/drug effects , Muscle, Smooth, Vascular/cytology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/physiology , Prostaglandins I/physiology , Pyridoxal Phosphate/analogs & derivatives , Pyridoxal Phosphate/pharmacology , Receptors, Purinergic P2/physiology , Suramin/pharmacology
10.
Am J Physiol Renal Physiol ; 289(2): F235-46, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16006589

ABSTRACT

The balance between vasodilator and vasoconstrictor pathways is key to the maintenance of homeostasis and the outcome of disease. In the kidney, prostaglandins (PGs) uphold this balance and regulate renal function: hemodynamics, renin secretion, growth responses, tubular transport processes, and cell fate. With the advent of cyclooxygenase (COX)-2-selective inhibitors, targeted deletions in mice (COX knockouts, PG receptor knockouts), and the discovery of intracrine signaling options for PGs (peroxisome proliferator-activated receptors and perinuclear PGE(2) receptors: EP(1,3,4)), many advances have been made in the study of arachidonic acid metabolites. Although prostacyclin (PGI(2)) is a major product of the COX pathway, there is very little emphasis on its importance to the kidney. This review will discuss PGI(2) biology and its relevance to different aspects of renal disease (growth, fibrosis, apoptosis), highlighting the most significant research from the past decade of PGI(2) literature, what we have learned from other organ systems, while stressing the significance of cross talk between various PGI(2) signaling pathways and its implications for renal health and disease.


Subject(s)
Kidney Diseases/physiopathology , Kidney/physiology , Prostaglandins I/physiology , Signal Transduction/physiology , Animals , Epoprostenol/biosynthesis , Epoprostenol/physiology , Health , Humans , Mice , Mice, Knockout , PPAR gamma/physiology , Receptors, Prostaglandin/metabolism
11.
Eur J Pharmacol ; 506(2): 151-6, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15588735

ABSTRACT

The aim of this study was to analyse the possible influence of endogenous prostacyclin on neuronal nitric oxide (NO) release induced by electrical field stimulation in mesenteric arteries from spontaneously hypertensive rats (SHR). Preincubation with the prostacyclin synthesis inhibitor tranylcypromine decreased NO release induced by electrical field stimulation, which was reversed by exogenous prostacyclin. Preincubation with tranylcypromine increased basal and electrical field stimulation-induced [3H]noradrenaline release. The nitric oxide synthase inhibitor Nomega-nitro-L-arginine methyl esther (L-NAME) increased the vasoconstrictor response induced by electrical field stimulation. In the presence of tranylcypromine, L-NAME did not modify the vasoconstrictor response induced by electrical field stimulation. In the presence of tranylcypromine and prostacyclin, LNAME increased the vasoconstrictor response to electrical field stimulation. These results indicate that endogenous prostacyclin positively modulates the neuronal NO release induced by electrical field stimulation and that this neuronal NO participates in the regulation of the vasomotor response.


Subject(s)
Mesenteric Arteries/metabolism , Neurons/metabolism , Nitric Oxide/metabolism , Prostaglandins I/physiology , Anesthetics, Local/pharmacology , Animals , Electric Stimulation , In Vitro Techniques , Male , Mesenteric Arteries/physiology , Monoamine Oxidase Inhibitors/pharmacology , Muscle Contraction/drug effects , Muscle Contraction/physiology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurons/physiology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Rats , Rats, Inbred SHR , Tetrodotoxin/pharmacology , Tranylcypromine/pharmacology
12.
Circulation ; 109(13): 1653-9, 2004 Apr 06.
Article in English | MEDLINE | ID: mdl-15023873

ABSTRACT

BACKGROUND: Microparticles (MPs) are membrane vesicles with procoagulant and proinflammatory properties released during cell activation. The present study was designed to dissect the effects evoked by T lymphocyte-derived MPs on vascular function. METHODS AND RESULTS: MPs were produced by treatment of the human lymphoid CEM T cell line with actinomycin D or phytohemagglutinin. Incubation of mouse aortic rings with 30 nmol/L MPs resulted in a time-dependent impairment of acetylcholine-induced relaxation of precontracted vessels, with a maximal reduction after 24 hours. MPs also impaired shear stress-induced dilatation of mouse small mesenteric arteries by affecting the nitric oxide (NO) and prostacyclin but not the endothelium-derived hyperpolarizing factor components of the response. However, neither alteration of calcium signaling in response to agonists nor reduction of cyclooxygenase-1 expression accounted for the impairment of the NO and prostacyclin components of the endothelial response. The effect of MPs was rather because of a decrease in expression of endothelial NO synthase and an overexpression of caveolin-1. Furthermore, lymphocyte-derived MPs from diabetic patients or in vivo circulating MPs from either diabetic or HIV-infected patients reduced endothelial NO synthase expression. Finally, the effects of MPs on endothelial cells were not driven through CD11a/CD18 adhesion molecules or the Fas/FasL pathway. CONCLUSIONS: MPs from T cells induce endothelial dysfunction in both conductance and resistance arteries by alteration of NO and prostacyclin pathways. MPs regulate protein expression for endothelial NO synthase and caveolin-1. These data contribute to a better understanding of the deleterious effects of enhanced circulating MPs observed in disorders with cardiovascular or immune complications.


Subject(s)
Cell Membrane/ultrastructure , Endothelium, Vascular/physiology , T-Lymphocytes/ultrastructure , Acetylcholine/pharmacology , Animals , Aorta/drug effects , Aorta/physiology , Biological Factors/physiology , Caveolin 1 , Caveolins/biosynthesis , Caveolins/genetics , Cell Membrane/chemistry , Cells, Cultured , Dactinomycin/pharmacology , Diabetes Mellitus/immunology , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Gene Expression Regulation , HIV Infections/immunology , Humans , In Vitro Techniques , Male , Membrane Lipids/chemistry , Mesenteric Arteries/physiology , Mice , Nitric Oxide/physiology , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/physiology , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Nitroarginine/pharmacology , Phosphatidylserines/chemistry , Phytohemagglutinins/pharmacology , Prostaglandins I/physiology , Stress, Mechanical , T-Lymphocytes/drug effects , Vasodilation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...