Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Chem Biol Interact ; 379: 110513, 2023 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-37116854

RESUMO

We investigated the effect of inhibition of 5-lipoxigenase (LOX) and 12-LOX pathways on the regeneration of skeletal muscle fibers after injury induced by a myotoxin (MTX) phospholipase A2 from snake venom in an in vivo experimental model. Gastrocnemius muscles of mice injected with MTX presented an increase in 5-LOX protein expression, while 12-LOX was found to be a constitutive protein of skeletal muscle. Animals that received oral treatments with 5-LOX inhibitor MK886 or 12-LOX inhibitor baicalein 30 min and 48 h after MTX-induced muscle injury showed a reduction in the inflammatory process characterized by a significant decrease of cell influx and injured fibers in the degenerative phase (6 and 24 h after injury). In the beginning of the regeneration process (3 days), mice that received MK886 showed fewer new basophilic fibers, suggesting fewer proliferative events and myogenic cell fusion. Furthermore, in the progression of tissue regeneration (14-21 days), the mice treated with 5-LOX inhibitor presented a lower quantity of central nucleus fibers and small-caliber fibers, culminating in a muscle that is more resistant to the stimulus of fatigue during muscle regeneration with a predominance of slow fibers. In contrast, animals early treated with the 12-LOX inhibitor presented functional fibers with higher diameters, less resistant to fatigue and predominance of fast heavy-chain myosin fibers as observed in control animals. These effects were accompanied by an earlier expression of myogenic factor MyoD. Our results suggest that both 5-LOX and 12-LOX pathways represent potential therapeutic targets for muscle regeneration. It appears that inhibition of the 5-LOX pathway represses only the degenerative process by reducing tissue inflammation levels. Meanwhile, inhibition of the 12-LOX pathway also favors the anticipation of maturation and earlier recovery of muscle fiber activity function after injury.


Assuntos
Araquidonato 12-Lipoxigenase , Doenças Musculares , Camundongos , Animais , Araquidonato 12-Lipoxigenase/farmacologia , Araquidonato 5-Lipoxigenase/farmacologia , Fibras Musculares Esqueléticas , Músculo Esquelético
2.
Circulation ; 147(19): 1444-1460, 2023 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-36987924

RESUMO

BACKGROUND: Myocardial ischemia-reperfusion (I/R) injury causes cardiac dysfunction to myocardial cell loss and fibrosis. Prevention of cell death is important to protect cardiac function after I/R injury. The process of reperfusion can lead to multiple types of cardiomyocyte death, including necrosis, apoptosis, autophagy, and ferroptosis. However, the time point at which the various modes of cell death occur after reperfusion injury and the mechanisms underlying ferroptosis regulation in cardiomyocytes are still unclear. METHODS: Using a left anterior descending coronary artery ligation mouse model, we sought to investigate the time point at which the various modes of cell death occur after reperfusion injury. To discover the key molecules involved in cardiomyocyte ferroptosis, we performed a metabolomics study. Loss/gain-of-function approaches were used to understand the role of 15-lipoxygenase (Alox15) and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (Pgc1α) in myocardial I/R injury. RESULTS: We found that apoptosis and necrosis occurred in the early phase of I/R injury, and that ferroptosis was the predominant form of cell death during the prolonged reperfusion. Metabolomic profiling of eicosanoids revealed that Alox15 metabolites accumulated in ferroptotic cardiomyocytes. We demonstrated that Alox15 expression was specifically increased in the injured area of the left ventricle below the suture and colocalized with cardiomyocytes. Furthermore, myocardial-specific knockout of Alox15 in mice alleviated I/R injury and restored cardiac function. 15-Hydroperoxyeicosatetraenoic acid (15-HpETE), an intermediate metabolite derived from arachidonic acid by Alox15, was identified as a trigger for cardiomyocyte ferroptosis. We explored the mechanism underlying its effects and found that 15-HpETE promoted the binding of Pgc1α to the ubiquitin ligase ring finger protein 34, leading to its ubiquitin-dependent degradation. Consequently, attenuated mitochondrial biogenesis and abnormal mitochondrial morphology were observed. ML351, a specific inhibitor of Alox15, increased the protein level of Pgc1α, inhibited cardiomyocyte ferroptosis, protected the injured myocardium, and caused cardiac function recovery. CONCLUSIONS: Together, our results established that Alox15/15-HpETE-mediated cardiomyocyte ferroptosis plays an important role in prolonged I/R injury.


Assuntos
Araquidonato 15-Lipoxigenase , Ferroptose , Traumatismo por Reperfusão Miocárdica , Animais , Camundongos , Apoptose , Araquidonato 12-Lipoxigenase/metabolismo , Araquidonato 12-Lipoxigenase/farmacologia , Araquidonato 15-Lipoxigenase/genética , Araquidonato 15-Lipoxigenase/metabolismo , Araquidonato 15-Lipoxigenase/farmacologia , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Necrose/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Ubiquitinas/metabolismo , Ubiquitinas/farmacologia
3.
Aging (Albany NY) ; 14(3): 1068-1086, 2022 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-35158337

RESUMO

Radiation therapy is a commonly used treatment modality for cancer. Although effective in providing local tumor control, radiation causes oxidative stress, inflammation, immunomodulatory and mitogenic cytokine production, extracellular matrix production, and premature senescence in lung parenchyma. The senescence associated secretory phenotype (SASP) can promote inflammation and stimulate alterations in the surrounding tissue. Therefore, we hypothesized that radiation-induced senescent parenchymal cells in irradiated lung would enhance tumor growth. Using a murine syngeneic tumor model of melanoma and non-small cell lung cancer lung metastasis, we demonstrate that radiation causes a significant increase in markers of premature senescence in lung parenchyma within 4 to 8 weeks. Further, injection of B16F0 (melanoma) or Lewis Lung carcinoma (epidermoid lung cancer) cells at these time points after radiation results in an increase in the number and size of pulmonary tumor nodules relative to unirradiated mice. Treatment of irradiated mice with a senolytic agent (ABT-737) or agents that prevent senescence (rapamycin, INK-128) was sufficient to reduce radiation-induced lung parenchymal senescence and to mitigate radiation-enhanced tumor growth. These agents abrogated radiation-induced expression of 12-Lipoxygenase (12-LOX), a molecule implicated in several deleterious effects of senescence. Deficiency of 12-LOX prevented radiation-enhanced tumor growth. Together, these data demonstrate the pro-tumorigenic role of radiation-induced senescence, introduces the dual TORC inhibitor INK-128 as an effective agent for prevention of radiation-induced normal tissue senescence, and identifies senescence-associated 12-LOX activity as an important component of the pro-tumorigenic irradiated tissue microenvironment. These studies suggest that combining senotherapeutic agents with radiotherapy may decrease post-therapy tumor growth.


Assuntos
Carcinoma Pulmonar de Lewis , Neoplasias Pulmonares , Melanoma Experimental , Animais , Araquidonato 12-Lipoxigenase/farmacologia , Carcinoma Pulmonar de Lewis/enzimologia , Carcinoma Pulmonar de Lewis/patologia , Processos de Crescimento Celular , Senescência Celular , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/patologia , Melanoma Experimental/enzimologia , Melanoma Experimental/patologia , Camundongos , Microambiente Tumoral
4.
Blood Adv ; 4(18): 4522-4537, 2020 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-32946570

RESUMO

Arterial thrombosis is the underlying cause for a number of cardiovascular-related events. Although dietary supplementation that includes polyunsaturated fatty acids (PUFAs) has been proposed to elicit cardiovascular protection, a mechanism for antithrombotic protection has not been well established. The current study sought to investigate whether an omega-6 essential fatty acid, docosapentaenoic acid (DPAn-6), and its oxidized lipid metabolites (oxylipins) provide direct cardiovascular protection through inhibition of platelet reactivity. Human and mouse blood and isolated platelets were treated with DPAn-6 and its 12-lipoxygenase (12-LOX)-derived oxylipins, 11-hydroxy-docosapentaenoic acid and 14-hydroxy-docosapentaenoic acid, to assess their ability to inhibit platelet activation. Pharmacological and genetic approaches were used to elucidate a role for DPA and its oxylipins in preventing platelet activation. DPAn-6 was found to be significantly increased in platelets following fatty acid supplementation, and it potently inhibited platelet activation through its 12-LOX-derived oxylipins. The inhibitory effects were selectively reversed through inhibition of the nuclear receptor peroxisome proliferator activator receptor-α (PPARα). PPARα binding was confirmed using a PPARα transcription reporter assay, as well as PPARα-/- mice. These approaches confirmed that selectivity of platelet inhibition was due to effects of DPA oxylipins acting through PPARα. Mice administered DPAn-6 or its oxylipins exhibited reduced thrombus formation following vessel injury, which was prevented in PPARα-/- mice. Hence, the current study demonstrates that DPAn-6 and its oxylipins potently and effectively inhibit platelet activation and thrombosis following a vascular injury. Platelet function is regulated, in part, through an oxylipin-induced PPARα-dependent manner, suggesting that targeting PPARα may represent an alternative strategy to treat thrombotic-related diseases.


Assuntos
Araquidonato 12-Lipoxigenase , Plaquetas , Animais , Araquidonato 12-Lipoxigenase/genética , Araquidonato 12-Lipoxigenase/farmacologia , Lipídeos , Camundongos , PPAR alfa/genética , PPAR alfa/farmacologia , Proliferadores de Peroxissomos/farmacologia
5.
Int J Mol Med ; 35(1): 195-201, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25395029

RESUMO

Peroxisome proliferator-activated receptor γ (PPARγ) expression and activity are increased in brain ischemic injury and its agonists have shown potential for brain injury protection. The influence of 12/15-lipoxygenase (12/15-LOX) on the activity of PPARγ in oxygen-glucose deprivation (OGD) and ischemia-reperfusion (I/R) was investigated. A middle cerebral artery occlusion/reperfusion model with Sprague Dawley (SD) rats was established. For I/R intervention, the rats were treated with the 12/15-LOX-derived product 12-hydroxyeicosatetraenoic acid (12-HETE) for 30 min before cerebral artery occlusion. Primary cortical neurons from SD rats were used to establish an OGD cell model. 12-HETE or a 12/15-LOX antisense oligonucleotide (asON-12/15-LOX) was added to OGD-treated neurons. Western blots, immunofluorescence and enzyme-linked immunosorbent assays detected protein. Reverse transcription-polymerase chain reaction analyzed the expression of the PPARγ target genes. PPARγ-DNA binding activity was determined by peroxisome proliferator responsive element luciferase reporter vectors. 12/15-LOX total protein increased significantly with I/R, and expression of 12-HETE was also upregulated. 12-HETE treatment increased PPARγ protein expression and inhibited inducible nitric oxide synthase protein expression, which was upregulated with I/R. PPARγ nuclear protein and 12/15-LOX total protein expression in OGD-treated neurons increased significantly. 12-HETE treatment increased the expression of PPARγ nuclear protein, upregulated the mRNA levels of PPARγ target genes (lipoprotein lipase and acyl-CoA oxidase) and enhanced PPARγ-DNA binding activity. asON-12/15-LOX treatment inhibited 12/15-LOX and PPARγ protein expression and lipoprotein lipase mRNA. Cerebral I/R injury in rats and OGD treatment in neurons promoted 12/15-LOX expression, and 12-HETE activated PPARγ. Therefore, PPARγ can be activated by the 12/15-LOX pathway during cerebral I/R injury.


Assuntos
Araquidonato 12-Lipoxigenase/metabolismo , Araquidonato 15-Lipoxigenase/metabolismo , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , PPAR gama/metabolismo , Traumatismo por Reperfusão/metabolismo , Animais , Araquidonato 12-Lipoxigenase/genética , Araquidonato 12-Lipoxigenase/farmacologia , Araquidonato 15-Lipoxigenase/genética , Araquidonato 15-Lipoxigenase/farmacologia , Córtex Cerebral/irrigação sanguínea , Modelos Animais de Doenças , Ativação Enzimática , Expressão Gênica , Glucose/metabolismo , Infarto da Artéria Cerebral Média/complicações , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Oxigênio/metabolismo , Ligação Proteica , Transporte Proteico , Ratos , Traumatismo por Reperfusão/etiologia
6.
Zhonghua Yi Xue Za Zhi ; 94(15): 1179-83, 2014 Apr 22.
Artigo em Chinês | MEDLINE | ID: mdl-24924720

RESUMO

OBJECTIVE: To explore the effects of 12/15-lipoxygenase antisense oligonucleotide (asON-12/15-LOX) on OGD (oxygen-glucose deprivation)-induced PPARγ (peroxisome proliferator-activated receptor γ) expression and nuclear translocation in primarily cultured cortical neurons. METHODS: After a 48-h pre-treatment of 12/15-LOX antisense oligonucleotide (asON), primarily cultured cortical neurons underwent 3-hour OGD followed by a 24-hour reperfusion.Immunofluorescent staining and Western blot were used to evaluate the expressions of 12/15-LOX and PPARγ as well as the nuclear translocation of PPARγ. RESULTS: Compared with the control group, the expressions of 12/15-LOX and PPARγ whole protein were enhanced in OGD group (t = -3.72 and -6.79, P = 0.03 and 0.04). And an increase of PPARγ in nucleus (t = -4.67, P = 0.02) could be noted with a simultaneous reduction in cytosol (t = 3.40, P = 0.04) after OGD, indicating an induction of nuclear translocation by OGD. Compared with OGD group, a pre-treatment of asON-12/15-LOX dramatically attenuated OGD-induced increase in 12/15-LOX whole protein expression (t = 5.03, P = 0.02). Compared with OGD group, a pre-treatment of asON-12/15-LOX greatly reduced OGD-induced increase in PPARγ total protein expression (t = 2.83, P = 0.04) and nuclear translocation (t = 7.05, P = 0.01 for nuclear protein; t = -5.47, P = 0.01 for cytosol protein). It indicated a possible link between 12/15-LOX and PPARγ. CONCLUSION: 12/15-LOX antisense oligonucleotide suppresses the expression and nuclear translocation of PPARγ in primarily cultured cortical neurons after OGD.


Assuntos
Araquidonato 12-Lipoxigenase/farmacologia , Araquidonato 15-Lipoxigenase/farmacologia , Neurônios/metabolismo , Oligonucleotídeos Antissenso/farmacologia , PPAR gama/metabolismo , Animais , Hipóxia Celular , Células Cultivadas , Córtex Cerebral/citologia , Meios de Cultura/química , Glucose/química , Neurônios/efeitos dos fármacos , Oxigênio/química , Ratos , Ratos Sprague-Dawley
7.
Thromb Res ; 125(4): e118-22, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19962724

RESUMO

INTRODUCTION: We have previously defined aspirin resistance detected by TEG PlateletMapping using arachidonic acid (AA). This aspirin resistance is observed as platelet activation (>20%) by AA in whole blood, even though the isolated platelets are inhibited by aspirin. This platelet activation in whole blood is due to a transcellular pathway mediated by platelets and leukocytes. METHODS: To determine if this PlateletMapping assay of aspirin resistance on pre-procedure blood samples correlated with an in vivo response we assayed the first voided urine samples collected 2-8 hours post interventional cardiology procedures for 11-dehydro thromboxane B2. RESULTS AND CONCLUSIONS: We detected 27 aspirin resistant patients out of a total of 81 (33%), in agreement with our previous study. All of these patients were on aspirin therapy, confirmed by a <20% aggregation response to AA by light transmission platelet aggregometry using isolated platelet rich plasma. Aspirin resistant patients urine samples (14 out of a total of 60 patients analyzed) contained significantly (P=0.008) higher 11-dehydro thromboxane B2 levels than the other 46 aspirin sensitive patients urine samples. Since our previous study implicated 12- and 15-lipoxygenases in this pathway, we also assayed for polymorphisms to determine any correlation with aspirin resistance. A correlation was found in a polymorphism affecting the lipoxygenase domain of platelet 12-lipoxygenase. This result indicates that aspirin resistance detected in whole blood by the TEG PlateletMapping assay correlates with a physiological consequence in terms of thromboxane formation. This is the first report of such a correlation.


Assuntos
Aspirina/farmacologia , Plaquetas/efeitos dos fármacos , Cardiologia/métodos , Ativação Plaquetária/efeitos dos fármacos , Tromboxano B2/análogos & derivados , Araquidonato 12-Lipoxigenase/farmacologia , Araquidonato 15-Lipoxigenase/farmacologia , Ácido Araquidônico/farmacologia , Plaquetas/fisiologia , Humanos , Ativação Plaquetária/fisiologia , Contagem de Plaquetas/métodos , Tromboxano B2/urina
8.
Arch Physiol Biochem ; 112(2): 119-29, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16931454

RESUMO

The expression and activity of the arachidonic acid-metabolizing enzyme leukocyte-type 12-lipoxygenase (12-LO) are augmented in cultured vascular endothelial and smooth muscle cells exposed to high glucose concentrations and in blood vessels of diabetic animals. The product of this enzyme, 12-hydroxyeicosatetraenoic acid (12-HETE), evokes two types of interactions in these cells: on one hand it acts as a pro-inflammatory factor that contributes to the initiation and progression of atherosclerotic lesions. Yet on the other, it protects the same cells against deleterious effects of high levels of intracellular glucose by downregulating the glucose transport system in the cells. In addition, it has been shown that 12-LO and 12-HETE support insulin-dependent glucose transporter-4 translocation to the plasma membrane by maintaining intact actin fiber network in the cardiomyocytes. Here we focus on the disparate cellular interactions by which 12-LO and 12-HETE affect the glucose transport system in vascular endothelial and smooth muscle cells and in cardiomyocytes.


Assuntos
Araquidonato 12-Lipoxigenase/farmacologia , Endotélio Vascular/efeitos dos fármacos , Ácidos Hidroxieicosatetraenoicos/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Células Cultivadas , Modelos Biológicos
9.
Thromb Haemost ; 85(6): 1037-42, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11434681

RESUMO

Megakaryocytic genes such as alphaIIbbeta3 can be expressed by malignant cells as part of the disturbances in their gene regulation. However, the function of the gene product greatly depends on the interaction of the ectopic protein with the new environment. The outside-in signaling of the ectopically expressed alphaIIbbeta3 integrin was studied in B16a murine melanoma cells using a monoclonal antibody, specifically directed to the activated conformation of alphaIIbbeta3, PAC-1 and the physiological ligand, fibrinogen. Ligation of alphaIIbbeta3 induced down-regulation of FAK but serine phosphorylation of three protein bands, 20/21, 85 and 140 kDa within 1-15 min. Flow cytometry indicated that the ligation of the receptor in B16a cells induces approximately 50% increase in phosphoserine positive cells within 5-15 min. 12-lipoxygenase is placed downstream in the signaling pathway, since ligation of alphaIIbbeta3 induces 12-HETE production within 5 min and pretreatment of tumor cells with select lipoxygenase inhibitior, Baicalein, prevents the increase in serine phosphorylation. Confocal microscopy of adherent tumor cells demonstrated rearrangement of actin filaments upon alphaIIbbeta3 ligation paralleled by downregulation of p125FAK and phoshotyrosine+ adhesion plaques and translocation of PKCalpha to stress fibers and cortical actin. PKC appears to be the major effector serine kinase of the alphaIIbbeta3-coupled signaling pathway, since pretreatment of tumor cells with a select PKC inhibitor, Calphostin C, prevents the ligation-induced serine phosphorylation. Previous studies have indicated a role for the 12-lipoxygenase-PKC signaling pathway in platelet aggregation as well as tumor invasion, therefore the involvement of this cascade in the signaling of the ectopic alphaIIbbeta3 integrin may partially explain its role in tumor progression.


Assuntos
Araquidonato 12-Lipoxigenase/farmacologia , Melanoma/patologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/fisiologia , Proteína Quinase C/farmacologia , Transdução de Sinais/fisiologia , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/análise , Animais , Anticorpos Monoclonais/farmacologia , Adesão Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Fibrinogênio/metabolismo , Citometria de Fluxo , Camundongos , Microscopia Confocal , Fosforilação/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/imunologia , Proteína Quinase C/antagonistas & inibidores , Serina/metabolismo , Células Tumorais Cultivadas
10.
Neuron ; 19(2): 453-63, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9292733

RESUMO

An early and highly specific decrease in glutathione (GSH) in the substantia nigra is associated with Parkinson's disease, and low levels of GSH lead to the degeneration of cultured dopaminergic neurons. Using immature cortical neurons and a clonal nerve cell line, it is shown that a decrease in GSH triggers the activation of neuronal 12-lipoxygenase (12-LOX), which leads to the production of peroxides, the influx of Ca2+, and ultimately to cell death. The supporting evidence includes: 1) inhibitors of arachidonate metabolism and 12-LOX block cell death induced by GSH depletion; 2) there is an increase in 12-LOX activity and a membrane translocation in HT22 cells, and an induction of the enzyme in primary cortical neurons following the reduction of GSH; 3) 12-LOX is directly inhibited by GSH; and 4) exogenous arachidonic acid potentiates cell death. These data show that the LOX pathway is a critical intermediate in at least some forms of neuronal degeneration.


Assuntos
Araquidonato 12-Lipoxigenase/farmacologia , Morte Celular/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Glutationa/farmacologia , Neurônios/efeitos dos fármacos , Animais , Linhagem Celular , Ácido Glutâmico/farmacologia , Ratos , Ratos Sprague-Dawley
11.
Am J Physiol ; 272(4 Pt 1): L573-9, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9142927

RESUMO

The effects of phosphonofluoridic acid, methyl-5,8,11,14-eicosatetraenyl ester (MAFP), a phospholipase A2 inhibitor, on pressor responses to angiotensin II (ANG II), norepinephrine (NE), serotonin (5-HT), the calcium channel opener BAY K 8644, and the thromboxane A2 mimic U-46619 were studied in the pulmonary vascular bed of the intact-chest cat. Under conditions of constant lobar blood flow, injections of ANG II, NE, 5-HT, U-46619, and BAY K 8644 into the lobar arterial perfusion circuit caused dose-related increases in lobar arterial pressure that were reproducible with respect to time. Infusion of MAFP into the perfused lobar artery at doses of 100 to 300 microg/kg for 10 min significantly reduced vasoconstrictor responses to ANG II; however, the phospholipase A2 inhibitor did not alter vasoconstrictor responses to BAY K 8644, 5-HT, NE, or U-46619. The combination of the higher dose of the phospholipase A2 inhibitor MAFP with the phospholipase C inhibitor U-73122 significantly affected vasoconstrictor responses to ANG II, NE, and 5-HT but not to BAY K 8644. In a separate series of animals, the effects of a lipoxygenase inhibitor, baicalein, were investigated. Infusion of baicalein into the perfused lobar artery at doses of 100 microg/kg for 10 min significantly reduced vasoconstrictor responses to ANG II but not the vasoconstrictor responses to BAY K 8644, 5-HT, NE, or U-46619. In a final series of experiments, the effects of a cyclooxygenase inhibitor, meclofenamate, were investigated, and intravenous injection of the meclofenamate at a dose of 2.5 mg/kg did not significantly affect vasoconstrictor responses to ANG II, 5-HT, BAY K 8644, NE, or U-46619. These data provide support for the hypothesis that pulmonary vasopressor responses to ANG II are mediated, in part, by the activation of phospholipase A2, phospholipase C, and the lipoxygenase pathway in the pulmonary vascular bed of the cat.


Assuntos
Araquidonato 12-Lipoxigenase/farmacologia , Inibidores de Ciclo-Oxigenase/farmacologia , Flavanonas , Fosfolipases A/farmacologia , Circulação Pulmonar/efeitos dos fármacos , Animais , Ácidos Araquidônicos/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Gatos , Combinação de Medicamentos , Inibidores Enzimáticos/farmacologia , Estrenos/farmacologia , Feminino , Flavonoides/farmacologia , Inibidores de Lipoxigenase/farmacologia , Masculino , Ácido Meclofenâmico/farmacologia , Organofosfonatos , Inibidores de Fosfodiesterase/farmacologia , Fosfolipases A/antagonistas & inibidores , Fosfolipases A2 , Pirrolidinonas/farmacologia , Fosfolipases Tipo C/antagonistas & inibidores , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia
12.
Br J Pharmacol ; 103(4): 1909-16, 1991 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-1655146

RESUMO

1. The addition of 2 x 10(8) human platelets to 8 x 10(6) polymorphonuclear leucocytes (PMNL) incubated in presence of 2.5 u ml-1 thrombin and 0.1 microM N-formyl-Met-Leu-Phe (FMLP) (or C5a or PAF) led to enhancement of leukotriene B4 (LTB4) synthesis by the PMNL (measured by h.p.l.c. as 20-hydroxy- and 20-carboxy-LTB4) from 4 +/- 1 pmol (in absence of platelets) to 26 +/- 4 pmol (mean +/- s.e.mean, n = 9). Platelets and thrombin were both essential for the enhancement of LTB4 synthesis. 2. Platelets also caused enhancement of LTB4 synthesis from (30 +/- 12 to 134 +/- 25 pmol, n = 6) when PMNL pretreated with granulocyte-macrophage colony-stimulating factor were used in similar experiments. 3. Enhancement of LTB4 synthesis was also observed (from 5 +/- 1.5 to 26.5 +/- 5 pmol, n = 9) when the supernatants of thrombin-activated platelet suspensions were added to FMLP-stimulated PMNL. 4. Supernatants of platelet suspensions activated by thrombin in presence of cyclo-oxygenase and 12-lipoxygenase inhibitors led to greater enhancement (from 5 +/- 3 to 153.5 +/- 27.5 pmol, n = 3) of LTB4 synthesis by FMLP-stimulated PMNL, suggesting that arachidonic acid itself, rather than its metabolites was responsible for the effects of platelets. 5. Addition of arachidonic acid to FMLP-stimulated PMNL at a concentration comparable to that measured in thrombin-activated platelet supernatants (0.2 +/- 0.025 microM, n = 6) mimicked the effect of platelets or platelet supernatants on LTB4 synthesis in FMLP-activated PMNL. 6. The present data indicate that under conditions of cell activation by physiological agonists, platelets can significantly increase the formation of the proinflammatory compound LTB4 in PMNL by providing arachidonic acid. These data lend support to the concept that platelet-PMNL interactions could modulate the inflammatory process.


Assuntos
Plaquetas/fisiologia , Leucotrieno B4/biossíntese , Neutrófilos/metabolismo , Trombina/fisiologia , Araquidonato 12-Lipoxigenase/farmacologia , Ácido Araquidônico/fisiologia , Humanos , Técnicas In Vitro , Leucotrieno B4/análise , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Fator de Ativação de Plaquetas/farmacologia , Ativação Plaquetária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...