Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
PLoS Pathog ; 15(6): e1007887, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31233552

RESUMEN

Leishmaniasis is a neglected tropical disease affecting millions of individuals worldwide. P2X7 receptor has been linked to the elimination of Leishmania amazonensis. Biological responses evoked by P2X7 receptor activation have been well-documented, including apoptosis, phagocytosis, cytokine release, such as IL-1ß. It was demonstrated that NLRP3 inflammasome activation and IL-1ß signaling participated in resistance against L. amazonensis. Furthermore, our group has shown that L. amazonensis elimination through P2X7 receptor activation depended on leukotriene B4 (LTB4) production and release. Therefore, we investigated whether L. amazonensis elimination by P2X7 receptor and LTB4 involved NLRP3 inflammasome activation and IL-1ß signaling. We showed that macrophages from NLRP3-/-, ASC-/-, Casp-1/11-/-, gp91phox-/- , and IL-1R-/- mice treated with ATP or LTB4 did not decrease parasitic load as was observed in WT mice. When ASC-/- macrophages were treated with exogenous IL-1ß, parasite killing was noted, however, we did not see parasitic load reduction in IL-1R-/- macrophages. Similarly, macrophages from P2X7 receptor-deficient mice treated with IL-1ß also showed decreased parasitic load. In addition, when we infected Casp-11-/- macrophages, neither ATP nor LTB4 were able to reduce parasitic load, and Casp-11-/- mice were more susceptible to L. amazonensis infection than were WT mice. Furthermore, P2X7-/- L. amazonensis-infected mice locally treated with exogenous LTB4 showed resistance to infection, characterized by lower parasite load and smaller lesions compared to untreated P2X7-/- mice. A similar observation was noted when infected P2X7-/- mice were treated with IL-1ß, i.e., lower parasite load and smaller lesions compared to P2X7-/- mice. These data suggested that L. amazonensis elimination mediated by P2X7 receptor and LTB4 was dependent on non-canonical NLRP3 inflammasome activation, ROS production, and IL-1ß signaling.


Asunto(s)
Inflamasomas/inmunología , Interleucina-1beta/inmunología , Leishmania/inmunología , Leishmaniasis/inmunología , Leucotrieno B4/inmunología , Macrófagos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Receptores Purinérgicos P2X7/inmunología , Transducción de Señal/inmunología , Animales , Inflamasomas/genética , Interleucina-1beta/genética , Leishmaniasis/genética , Leishmaniasis/patología , Leucotrieno B4/genética , Macrófagos/parasitología , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Receptores Purinérgicos P2X7/genética , Transducción de Señal/genética
2.
Arterioscler Thromb Vasc Biol ; 39(6): 1191-1202, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30943774

RESUMEN

Objective- To determine whether pulmonary arterial hypertension is associated with endothelial cell (EC)-Cav-1 (caveolin-1) depletion, EC-derived extracellular vesicle cross talk with macrophages, and proliferation of Cav-1 depleted ECs via TGF-ß (transforming growth factor-ß) signaling. Approach and Results- Pulmonary vascular disease was induced in Sprague-Dawley rats by exposure to a single injection of VEGFRII (vascular endothelial growth factor receptor II) antagonist SU5416 (Su) followed by hypoxia (Hx) plus normoxia (4 weeks each-HxSu model) and in WT (wild type; Tie2.Cre-; Cav1 lox/lox) and EC- Cav1-/- (Tie2.Cre+; Cav1 fl/fl) mice (Hx: 4 weeks). We observed reduced lung Cav-1 expression in the HxSu rat model in association with increased Cav-1+ extracellular vesicle shedding into the circulation. Whereas WT mice exposed to hypoxia exhibited increased right ventricular systolic pressure and pulmonary microvascular thickening compared with the group maintained in normoxia, the remodeling was further increased in EC- Cav1-/- mice indicating EC Cav-1 expression protects against hypoxia-induced pulmonary hypertension. Depletion of EC Cav-1 was associated with reduced BMPRII (bone morphogenetic protein receptor II) expression, increased macrophage-dependent TGF-ß production, and activation of pSMAD2/3 signaling in the lung. In vitro, in the absence of Cav-1, eNOS (endothelial NO synthase) dysfunction was implicated in the mechanism of EC phenotype switching. Finally, reduced expression of EC Cav-1 in lung histological sections from human pulmonary arterial hypertension donors was associated with increased plasma concentration of Cav-1, extracellular vesicles, and TGF-ß, indicating Cav-1 may be a plasma biomarker of vascular injury and key determinant of TGF-ß-induced pulmonary vascular remodeling. Conclusions- EC Cav-1 depletion occurs, in part, via Cav-1+ extracellular vesicle shedding into the circulation, which contributes to increased TGF-ß signaling, EC proliferation, vascular remodeling, and pulmonary arterial hypertension.


Asunto(s)
Caveolina 1/deficiencia , Células Endoteliales/metabolismo , Vesículas Extracelulares/metabolismo , Hipertensión Arterial Pulmonar/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Remodelación Vascular , Adolescente , Adulto , Anciano , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Estudios de Casos y Controles , Caveolina 1/genética , Proliferación Celular , Modelos Animales de Enfermedad , Células Endoteliales/patología , Vesículas Extracelulares/patología , Femenino , Humanos , Hipoxia/complicaciones , Indoles , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Hipertensión Arterial Pulmonar/etiología , Hipertensión Arterial Pulmonar/patología , Pirroles , Ratas Sprague-Dawley , Transducción de Señal , Proteínas Smad/metabolismo , Adulto Joven
3.
J Hepatol ; 67(4): 716-726, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28554875

RESUMEN

BACKGROUND & AIMS: The severity of sepsis can be linked to excessive inflammatory responses resulting in hepatic injury. P2X7 receptor activation by extracellular ATP (eATP) exacerbates inflammation by augmenting cytokine production; while CD39 (ENTPD1) scavenges eATP to generate adenosine, thereby limiting P2X7 activation and resulting in A2A receptor stimulation. We aim to determine how the functional interaction of P2X7 receptor and CD39 control the macrophage response, and consequently impact on sepsis and liver injury. METHODS: Sepsis was induced by cecal ligation and puncture in C57BL/6 wild-type (WT) and CD39-/- mice. Several in vitro assays were performed using peritoneal or bone marrow derived macrophages to determine CD39 ectonucleotidase activity and its role in sepsis-induced liver injury. RESULTS: CD39 expression in macrophages limits ATP-P2X7 receptor pro-inflammatory signaling. P2X7 receptor paradoxically boosts CD39 activity. Inhibition and/or deletion of P2X7 receptor in LPS-primed macrophages attenuates cytokine production and inflammatory signaling as well as preventing ATP-induced increases in CD39 activity. Septic CD39-/- mice exhibit higher levels of inflammatory cytokines and show more pronounced liver injury than WT mice. Pharmacological P2X7 blockade largely prevents tissue damage, cell apoptosis, cytokine production, and the activation of inflammatory signaling pathways in the liver from septic WT, while only attenuating these outcomes in CD39-/- mice. Furthermore, the combination of P2X7 blockade with adenosine A2A receptor stimulation completely inhibits cytokine production, the activation of inflammatory signaling pathways, and protects septic CD39-/- mice against liver injury. CONCLUSIONS: CD39 attenuates sepsis-associated liver injury by scavenging eATP and ultimately generating adenosine. We propose boosting of CD39 would suppress P2X7 responses and trigger adenosinergic signaling to limit systemic inflammation and restore liver homeostasis during the acute phase of sepsis. Lay summary: CD39 expression in macrophages limits P2X7-mediated pro-inflammatory responses, scavenging extracellular ATP and ultimately generating adenosine. CD39 genetic deletion exacerbates sepsis-induced experimental liver injury. Combinations of a P2X7 antagonist and adenosine A2A receptor agonist are hepatoprotective during the acute phase of abdominal sepsis.


Asunto(s)
Antígenos CD/metabolismo , Apirasa/metabolismo , Hígado/inmunología , Hígado/lesiones , Receptores Purinérgicos P2X7/metabolismo , Sepsis/inmunología , Agonistas del Receptor de Adenosina A2/farmacología , Adenosina Trifosfato/metabolismo , Animales , Antígenos CD/genética , Apirasa/deficiencia , Apirasa/genética , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Interleucina-1beta/biosíntesis , Hígado/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Antagonistas del Receptor Purinérgico P2X/farmacología , Receptores Purinérgicos P2X7/deficiencia , Receptores Purinérgicos P2X7/genética , Factor de Transcripción STAT3/metabolismo , Sepsis/terapia , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
4.
Front Immunol ; 14: 1254762, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37908354

RESUMEN

Schistosomiasis-associated Pulmonary Arterial Hypertension (Sch-PAH) is a life-threatening complication of chronic S. mansoni infection that can lead to heart failure and death. During PAH, the expansion of apoptosis-resistant endothelial cells (ECs) has been extensively reported; however, therapeutic approaches to prevent the progression or reversal of this pathological phenotype remain clinically challenging. Previously, we showed that depletion of the anti-apoptotic protein Caveolin-1 (Cav-1) by shedding extracellular vesicles contributes to shifting endoprotective bone morphogenetic protein receptor 2 (BMPR2) towards transforming growth factor beta (TGF-ß)-mediated survival of an abnormal EC phenotype. However, the mechanism underlying the reduced endoprotection in PAH remains unclear. Interestingly, recent findings indicate that, similar to the gut, healthy human lungs are populated by diverse microbiota, and their composition depends significantly on intrinsic and extrinsic host factors, including infection. Despite the current knowledge that the disruption of the gut microbiome contributes to the development of PAH, the role of the lung microbiome remains unclear. Thus, using a preclinical animal model of Sch-PAH, we tested whether S. mansoni infection alters the gut-lung microbiome composition and causes EC injury, initiating the expansion of an abnormal EC phenotype observed in PAH. Indeed, in vivo stimulation with S. mansoni eggs significantly altered the gut-lung microbiome profile, in addition to promoting injury to the lung vasculature, characterized by increased apoptotic markers and loss of endoprotective expression of lung Cav-1 and BMPR2. Moreover, S. mansoni egg stimulus induced severe pulmonary vascular remodeling, leading to elevated right ventricular systolic pressure and hypertrophy, characteristic of PAH. In vitro, exposure to the immunodominant S. mansoni egg antigen p40 activated TLR4/CD14-mediated transient phosphorylation of Cav-1 at Tyr14 in human lung microvascular EC (HMVEC-L), culminating in a mild reduction of Cav-1 expression, but failed to promote death and shedding of extracellular vesicles observed in vivo. Altogether, these data suggest that disruption of the host-associated gut-lung microbiota may be essential for the emergence and expansion of the abnormal lung endothelial phenotype observed in PAH, in addition to S. mansoni eggs and antigens.


Asunto(s)
Microbioma Gastrointestinal , Hipertensión Pulmonar , Hipertensión Arterial Pulmonar , Esquistosomiasis , Animales , Ratones , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Caveolina 1/genética , Células Endoteliales/metabolismo , Hipertensión Pulmonar/etiología , Pulmón/patología , Hipertensión Arterial Pulmonar/etiología , Hipertensión Arterial Pulmonar/patología , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Esquistosomiasis/metabolismo
5.
Nat Prod Res ; 35(22): 4870-4875, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32174171

RESUMEN

We investigated the role of triterpene barbinervic acid from Eugenia punicifolia dichloromethane extract in vasopressor responses. Renal arteries were cannulated and perfused with Krebs-Hepes solution. Changes in aorta isometric tension were recorded and transferred to a data acquisition system. Cumulative curves were constructed based on the maximum effect of agonists. Barbinervic acid reduced the renal tonus induced by NA in a NO-dependent manner (IC50 = 30 µM). Triterpene (70 µM) also induced rapid and transient relaxation in aorta that had been precontracted with K+ (53.2 ± 0.05%) or phenylephrine (36.7 ± 0.05%). In silico data revealed two possible active sites for interactions between barbinervic acid and NO synthase. Barbinervic acid showed a vasodilator effect and could potentially be used as a template for developing new molecules for the treatment of cardiovascular disease.


Asunto(s)
Eugenia , Triterpenos , Simulación por Computador , Extractos Vegetales/farmacología , Hojas de la Planta , Triterpenos/farmacología
6.
Eur J Pharmacol ; 525(1-3): 54-9, 2005 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-16289527

RESUMEN

The activity and protein expression of plasma membrane and sarco(endo)plasmic reticulum (Ca2+-Mg2+)ATPases and ryanodine receptors were investigated in surgically denervated rat vas deferens. The function of thapsigargin-sensitive but not thapsigargin-resistant (Ca2+-Mg2+)ATPase (from sarco(endo)plasmic reticulum and plasma membrane, respectively), evidenced by enzyme activity and Ca2+ uptake experiments, was significantly depressed by 30-50% when compared to innervated vas. Western blots showed that such reduction in sarco(endo)plasmic reticulum (Ca2+-Mg2+)ATPase performance was accompanied by a decrement of similar magnitude in sarco(endo)plasmic reticulum (Ca2+-Mg2+)ATPase type 2 protein expression, without any significant change in plasma membrane (Ca2+-Mg2+)ATPase expression. Finally, [3H]ryanodine binding revealed that the density of ryanodine binding sites was reduced by 45% after denervation without modification in affinity. The present findings demonstrate that sarco(endo)plasmic reticulum proteins involved in intracellular calcium homeostasis are clearly down-regulated and brings further evidence of a modified calcium translocation in denervated rat vas deferens.


Asunto(s)
ATPasas Transportadoras de Calcio/antagonistas & inhibidores , Calcio/metabolismo , Retículo Sarcoplasmático/enzimología , Conducto Deferente/inervación , Animales , ATPasa de Ca(2+) y Mg(2+)/metabolismo , Membrana Celular/enzimología , Membrana Celular/metabolismo , Desnervación , Homeostasis , Masculino , Ratas , Ratas Wistar , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico , Conducto Deferente/enzimología , Conducto Deferente/metabolismo
7.
Toxicon ; 67: 55-62, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23474269

RESUMEN

In this work we evaluated the ability of suramin, a polysulfonated naphthylurea derivative, to antagonize the cytotoxic and enzymatic effects of the crude venom of Apis mellifera. Suramin was efficient to decrease the lethality in a dose-dependent way. The hemoconcentration caused by lethal dose injection of bee venom was abolished by suramin (30 µg/g). The edematogenic activity of the venom (0.3 µg/g) was antagonized by suramin (10 µg/g) in all treatment protocols. The changes in the vascular permeability caused by A. mellifera (1 µg/g) venom were inhibited by suramin (30 µg/g) in the pre- and posttreatment as well as when the venom was preincubated with suramin. In addition, suramin also inhibited cultured endothelial cell lesion, as well as in vitro myotoxicity, evaluated in mouse extensor digitorum longus muscle, which was inhibited by suramin (10 and 25 µM), decreasing the rate of CK release, showing that suramin protected the sarcolemma against damage induced by components of bee venom (2.5 µg/mL). Moreover, suramin inhibited the in vivo myotoxicity induced by i.m. injection of A. mellifera venom in mice (0.5 µg/g). The analysis of the area under the plasma CK vs. time curve showed that preincubation, pre- and posttreatment with suramin (30 µg/g) inhibited bee venom myotoxic activity in mice by about 89%, 45% and 40%, respectively. Suramin markedly inhibited the PLA2 activity in a concentration-dependent way (1-30 µM). Being suramin a polyanion molecule, the effects observed may be due to the interaction of its charges with the polycation components present in A. mellifera bee venom.


Asunto(s)
Antivenenos/farmacología , Venenos de Abeja/farmacología , Fibras Musculares Esqueléticas/efectos de los fármacos , Suramina/farmacología , Animales , Venenos de Abeja/antagonistas & inhibidores , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Creatina Quinasa/sangre , Edema/inducido químicamente , Edema/tratamiento farmacológico , Edema/patología , Endotelio Vascular/efectos de los fármacos , Eritrocitos/efectos de los fármacos , Azul de Evans , Hematócrito , Inyecciones Intramusculares , Longevidad/efectos de los fármacos , Masculino , Ratones , Contracción Muscular/efectos de los fármacos , Fibras Musculares Esqueléticas/patología , Fosfolipasas A2/metabolismo , Ratas , Sarcolema/efectos de los fármacos , Sarcolema/enzimología , Piel/irrigación sanguínea
8.
PLoS One ; 6(8): e23547, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21853150

RESUMEN

BACKGROUND AND AIMS: Schistosomiasis is an intravascular parasitic disease associated with inflammation. Endothelial cells control leukocyte transmigration and vascular permeability being modulated by pro-inflammatory mediators. Recent data have shown that endothelial cells primed in vivo in the course of a disease keep the information in culture. Herein, we evaluated the impact of schistosomiasis on endothelial cell-regulated events in vivo and in vitro. METHODOLOGY AND PRINCIPAL FINDINGS: The experimental groups consisted of Schistosoma mansoni-infected and age-matched control mice. In vivo infection caused a marked influx of leukocytes and an increased protein leakage in the peritoneal cavity, characterizing an inflamed vascular and cellular profile. In vitro leukocyte-mesenteric endothelial cell adhesion was higher in cultured cells from infected mice as compared to controls, either in the basal condition or after treatment with the pro-inflammatory cytokine tumor necrosis factor (TNF). Nitric oxide (NO) donation reduced leukocyte adhesion to endothelial cells from control and infected groups; however, in the later group the effect was more pronounced, probably due to a reduced NO production. Inhibition of control endothelial NO synthase (eNOS) increased leukocyte adhesion to a level similar to the one observed in the infected group. Besides, the adhesion of control leukocytes to endothelial cells from infected animals is similar to the result of infected animals, confirming that schistosomiasis alters endothelial cells function. Furthermore, NO production as well as the expression of eNOS were reduced in cultured endothelial cells from infected animals. On the other hand, the expression of its repressor protein, namely caveolin-1, was similar in both control and infected groups. CONCLUSION/SIGNIFICANCE: Schistosomiasis increases vascular permeability and endothelial cell-leukocyte interaction in vivo and in vitro. These effects are partially explained by a reduced eNOS expression. In addition, our data show that the disease primes endothelial cells in vivo, which keep the acquired phenotype in culture.


Asunto(s)
Comunicación Celular , Células Endoteliales/patología , Leucocitos/patología , Esquistosomiasis/patología , Animales , Caveolina 1/metabolismo , Adhesión Celular , Movimiento Celular , Células Cultivadas , Células Endoteliales/enzimología , Masculino , Mesenterio/patología , Ratones , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo III/metabolismo , Cavidad Peritoneal/patología , Schistosoma mansoni/fisiología , Esquistosomiasis/enzimología
9.
Eur J Med Chem ; 46(7): 3000-12, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21549456

RESUMEN

We described herein the discovery of 1-(2-(benzo[d] [1,3]dioxol-6-yl)ethyl)-4-(2-methoxyphenyl) piperazine (LASSBio-772), as a novel potent and selective alpha 1A/1D adrenoceptor (AR) antagonist selected after screening of functionalized N-phenylpiperazine derivatives in phenylephrine-induced vasoconstriction of rabbit aorta rings. The affinity of LASSBio-772 for alpha 1A and alpha 1B AR subtypes was determined through displacement of [(3)H]prazosin binding. We obtained Ki values of 0.14 nM for the alpha 1A-AR, similar to that displayed by tamsulosin (K(i) = 0.13 nM) and 5.55 nM for the alpha 1B-AR, representing a 40-fold higher affinity for alpha 1A-AR. LASSBio-772 also presented high affinity (K(B) = 0.025 nM) for the alpha 1D-AR subtype in the functional rat aorta assay, showing to be equipotent to tamsulosin (K(B) = 0.017 nM).


Asunto(s)
Antagonistas de Receptores Adrenérgicos alfa 1/síntesis química , Aorta/efectos de los fármacos , Benzodioxoles/síntesis química , Membrana Celular/efectos de los fármacos , Piperazinas/síntesis química , Receptores Adrenérgicos alfa 1/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Animales , Benzodioxoles/farmacología , Membrana Celular/metabolismo , Hepatocitos/química , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Hígado/citología , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Fenilefrina/farmacología , Piperazinas/farmacología , Prazosina/metabolismo , Prazosina/farmacología , Unión Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Conejos , Ratas , Receptores Adrenérgicos alfa 1/química , Sulfonamidas/farmacología , Tamsulosina , Técnicas de Cultivo de Tejidos , Tritio , Vasoconstricción/efectos de los fármacos
10.
Pharmacol Ther ; 126(3): 251-62, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20398699

RESUMEN

Melatonin, the darkness hormone, synchronizes several physiological functions to light/dark cycle. Besides the awake/sleep cycle that is intuitively linked to day/night, daily variations in memory acquisition and innate or acquired immune responses are some of the major activities linked to melatonin rhythm. The daily variation of these complex processes is due to changes in specific mechanisms. In the last years we focused on the influence of melatonin on the expression and function of nicotinic acetylcholine receptors (nAChRs). Melatonin, either "in vivo" or "in vitro", increases, in a selective manner, the efficiency of alpha-bungarotoxin (alpha-BTX)-sensitive nAChRs. Melatonin's effect on receptors located in rat sympathetic nerve terminals, cerebellum, skeletal muscle and chick retina, was tested. We observed that melatonin is essential for the development of alpha-BTX-sensitive nAChRs, and important for receptor maintenance in aging models. Taking into account that both melatonin and alpha-7 nAChRs (one of the subtypes sensitive to alpha-BTX) are involved in the development of Alzheimer's disease, here we discuss the possibility of a therapeutic strategy focused on both melatonin replacement and its potential association with cholinergic drugs.


Asunto(s)
Colinérgicos/administración & dosificación , Melatonina/administración & dosificación , Melatonina/fisiología , Receptores Nicotínicos/fisiología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Animales , Quimioterapia Combinada , Humanos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
11.
J Pineal Res ; 41(3): 267-74, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16948788

RESUMEN

Endothelial cell function is a major player on the regulation of both vascular tonus and permeability. Activation of nitric oxide synthase (NOS) by bradykinin is one physiological pathway for the well-known vascular relaxation mediated by endothelial-derived nitric oxide (NO). In this study we investigated if melatonin, which is known to modulate endothelial cell function and NO production in other tissues, is able to impair bradykinin-induced NO production in vitro. Rat microvascular endothelial cells were incubated with fluorescent dyes to detect either NO or Ca2+. In addition, cGMP levels were measured by enzyme immunoassay. We found that while bradykinin (1-100 nm) increased both cytosolic Ca2+ and NO production, melatonin (1 nm) abolished this NO production but not cytosolic Ca2+ elevation. N-acetylserotonin (0.1 and 1 nm) had the same effect, while the selective agonist for MT3 receptors (5-MCA-NAT, 1 nm) had no effect. Moreover, nonselective and MT2-selective antagonists did not alter the effect of melatonin, suggesting that it is not mediated by MT melatonin receptors. A possible direct inhibition of calmodulin was also discarded as melatonin did not mimic the effect of calmidazolium on cytosolic Ca2+. Melatonin also abolished cGMP production induced by 1 microm bradykinin, indicating that the NO downstream effect is impaired. Thus, here we show that melatonin reduces NO production induced by bradykinin by a mechanism upstream to the interaction of Ca2+ -calmodulin with NOS. Moreover, this effect might be the basis of the diurnal variation in endothelial cell function.


Asunto(s)
Endotelio Vascular/metabolismo , Melatonina/fisiología , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Animales , Células Cultivadas , Células Endoteliales/metabolismo , Masculino , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA