Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Transl Med ; 20(1): 590, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36514072

RESUMEN

BACKGROUND AND AIMS: Kinin B1 receptors (B1Rs) are implicated in the pathogenesis of fibrosis. This study examined the anti-fibrotic effects of B1R blockade with BI 113823 in two established mouse models of hepatic fibrosis induced by intraperitoneal carbon tetrachloride (CCl4) injection or bile duct ligation (BDL). The mechanisms underlying the protection afforded by B1R inhibition were examined using human peripheral blood cells and LX2 human hepatic stellate cells (HSCs). METHODS: Fibrotic liver diseases were induced in mice by intraperitoneal carbon tetrachloride (CCl4) injection for 6 weeks, and by bile duct ligation (BDL) for 3 weeks, respectively. Mice received daily treatment of vehicle or BI 113823 (B1R antagonist) from onset of the experiment until the end of the study. RESULTS: B1Rs were strongly induced in fibrotic mouse liver. BI 113823 significantly attenuated liver fibrosis and portal hypertension (PH), and improved survival in both CCl4 and BDL mice. BI 113823 significantly reduced the expression of fibrotic proteins α-SMA, collagens 1, 3, 4, and profibrotic growth factors PDGF, TGFß, CTGF, VEGF, proliferating cell nuclear antigen; and reduced hepatic Akt phosphorylation in CCl4- and BDL-induced liver fibrosis. BI 113823 also reduced expression of Cytokines IL-1, IL-6; chemokines MCP-1, MCP-3 and infiltration of inflammatory cells; and inhibited human monocyte and neutrophil activation, transmigration, TNF-α & MPO production in vitro. BI 113823 inhibited TGF-ß and B1R agonist-stimulated human-HSC activation, contraction, proliferation, migration and fibrosis protein expression, and inhibited activation of PI3K/Akt signalling pathway. CONCLUSIONS: B1Rs merits consideration as a novel therapeutic target for chronic liver fibrosis and PH.


Asunto(s)
Hipertensión Portal , Cirrosis Hepática , Receptores de Péptidos , Animales , Humanos , Ratones , Tetracloruro de Carbono , Fibrosis , Células Estrelladas Hepáticas , Hipertensión Portal/complicaciones , Hipertensión Portal/tratamiento farmacológico , Hipertensión Portal/metabolismo , Cininas/metabolismo , Cininas/farmacología , Cininas/uso terapéutico , Hígado/patología , Cirrosis Hepática/complicaciones , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Receptores de Péptidos/antagonistas & inhibidores
2.
J Transl Med ; 19(1): 340, 2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34372885

RESUMEN

BACKGROUND: To examine the effects of BI 1029539 (GS-248), a novel selective human microsomal prostaglandin E synthase-1 (mPGES-1) inhibitor, in experimental models of acute lung injury (ALI) and sepsis in transgenic mice constitutively expressing the mPGES1 (Ptges) humanized allele. METHODS: Series 1: Lipopolysaccharide (LPS)-induced ALI. Mice were randomized to receive vehicle, BI 1029539, or celecoxib. Series 2: Cecal ligation and puncture-induced sepsis. Mice were randomized to receive vehicle or BI 1029539. RESULTS: Series 1: BI 1029539 or celecoxib reduced LPS-induced lung injury, with reduction in neutrophil influx, protein content, TNF-ɑ, IL-1ß and PGE2 levels in bronchoalveolar lavage (BAL), myeloperoxidase activity, expression of mPGES-1, cyclooxygenase (COX)-2 and intracellular adhesion molecule in lung tissue compared with vehicle-treated mice. Notably, prostacyclin (PGI2) BAL concentration was only lowered in celecoxib-treated mice. Series 2: BI 1029539 significantly reduced sepsis-induced BAL inflammatory cell recruitment, lung injury score and lung expression of mPGES-1 and inducible nitric oxide synthase. Treatment with BI 1029539 also significantly prolonged survival of mice with severe sepsis. Anti-inflammatory and anti-migratory effect of BI 1029539 was confirmed in peripheral blood leukocytes from healthy volunteers. CONCLUSIONS: BI 1029539 ameliorates leukocyte infiltration and lung injury resulting from both endotoxin-induced and sepsis-induced lung injury.


Asunto(s)
Lesión Pulmonar Aguda , Sepsis , Lesión Pulmonar Aguda/tratamiento farmacológico , Animales , Ciclooxigenasa 2/metabolismo , Dinoprostona , Modelos Animales de Enfermedad , Humanos , Pulmón/metabolismo , Ratones , Óxido Nítrico Sintasa de Tipo II , Prostaglandina-E Sintasas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
3.
Respir Res ; 22(1): 281, 2021 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-34717626

RESUMEN

BACKGROUND: This study examined whether BI113823, a novel selective kinin B1 receptor antagonist can reverse established pulmonary arterial hypertension (PAH), prevent right heart failure and death, which is critical for clinical translation. METHODS: Left pneumonectomized male Wistar rats were injected with monocrotaline to induce PAH. Three weeks later, when PAH was well established, the rats received daily treatment of BI113823 or vehicle for 3 weeks. RESULTS: Treatment with BI113823 from day 21 to day 42 after monocrotaline injection reversed established PAH as shown by normalized values of mean pulmonary arterial pressure (mPAP). BI113823 therapy reversed pulmonary vascular remodeling, pulmonary arterial neointimal formation, and heart and lung fibrosis, reduced right ventricular pressure, right heart hypertrophy, improved cardiac output, and prevented right heart failure and death. Treatment with BI113823 reduced TNF-α and IL-1ß, and macrophages recruitment in bronchoalveolar lavage, reduced CD-68 positive macrophages and expression of proliferating cell nuclear antigen (PCNA) in the perivascular areas, and reduced expression of iNOS, B1 receptors, matrix metalloproteinase (MMP)-2 and MMP-9 proteins, and the phosphorylation of ERK1/2 and AKT in lung. Treatment with BI113823 reduced mRNA expression of ANP, BNP, ßMHC, CGTF, collange-I and IV in right heart, compared to vehicle treated controls. In human monocytes cultures, BI113823 reduced LPS-induced TNF-α production, MMP-2 and MMP-9 expression, and reduced TNF-α-induced monocyte migration. CONCLUSIONS: We conclude that BI113823 reverses preexisting severe experimental pulmonary hypertension via inhibition of macrophage infiltration, cytokine production, as well as down regulation of matrix metalloproteinase proteins.


Asunto(s)
Cininas/antagonistas & inhibidores , Neointima/patología , Hipertensión Arterial Pulmonar/patología , Arteria Pulmonar/patología , Túnica Íntima/patología , Remodelación Vascular/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Humanos , Masculino , Hipertensión Arterial Pulmonar/tratamiento farmacológico , Hipertensión Arterial Pulmonar/metabolismo , Arteria Pulmonar/efectos de los fármacos , Ratas , Ratas Wistar , Túnica Íntima/efectos de los fármacos
4.
FASEB J ; 33(12): 13966-13981, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31638830

RESUMEN

The cause of antiseizure drug (ASD) resistance in epilepsy is poorly understood. Here, we focus on the transporter P-glycoprotein (P-gp) that is partly responsible for limited ASD brain uptake, which is thought to contribute to ASD resistance. We previously demonstrated that cyclooxygenase-2 (COX-2) and the prostaglandin E receptor, prostanoid E receptor subtype 1, are involved in seizure-mediated P-gp up-regulation. Thus, we hypothesized that inhibiting microsomal prostaglandin E2 (PGE2) synthase-1 (mPGES-1), the enzyme generating PGE2, prevents blood-brain barrier P-gp up-regulation after status epilepticus (SE). To test our hypothesis, we exposed isolated brain capillaries to glutamate ex vivo and used a combined in vivo-ex vivo approach by isolating brain capillaries from humanized mPGES-1 mice to study P-gp levels. We demonstrate that glutamate signaling through the NMDA receptor, cytosolic phospholipase A2, COX-2, and mPGES-1 increases P-gp protein expression and transport activity levels. We show that mPGES-1 is expressed in human, rat, and mouse brain capillaries. We show that BI1029539, an mPGES-1 inhibitor, prevented up-regulation of P-gp expression and transport activity in capillaries exposed to glutamate and in capillaries from humanized mPGES-1 mice after SE. Our data provide key signaling steps underlying seizure-induced P-gp up-regulation and suggest that mPGES-1 inhibitors could potentially prevent P-gp up-regulation in epilepsy.-Soldner, E. L. B., Hartz, A. M. S., Akanuma, S.-I., Pekcec, A., Doods, H., Kryscio, R. J., Hosoya, K.-I., Bauer, B. Inhibition of human microsomal PGE2 synthase-1 reduces seizure-induced increases of P-glycoprotein expression and activity at the blood-brain barrier.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Barrera Hematoencefálica/metabolismo , Dinoprostona/metabolismo , Microsomas/metabolismo , Prostaglandina-E Sintasas/metabolismo , Convulsiones/metabolismo , Animales , Transporte Biológico/fisiología , Encéfalo/metabolismo , Capilares/metabolismo , Ciclooxigenasa 2/metabolismo , Epilepsia/metabolismo , Femenino , Ácido Glutámico/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología
5.
J Infect Dis ; 213(4): 532-40, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26310310

RESUMEN

BACKGROUND: This study examined the therapeutic effects of an orally active nonpeptide kinin B1 receptor antagonist, BI113823, in a clinically relevant experimental model of polymicrobial sepsis in rats. METHODS: Sepsis was induced by cecal ligation and puncture (CLP). Animals received treatment with either vehicle or BI113823. The experiment was terminated in the first set of animals 15 hours after CLP. Seven-day survival following CLP was determined in the second set of animals. RESULTS: Compared with vehicle treatment, administration of BI113823 reduced neutrophil and macrophage infiltration, reduced cytokine production, attenuated intestinal mucosal hyperpermeability, prevented hemodynamic derangement, and improved cardiac output. Furthermore, administration of BI113823 reduced inducible nitric oxide synthase expression and the injury score in the lung and attenuated nuclear factor ĸB activation and apoptosis in the liver. Treatment with BI113823 also reduced plasma levels of cardiac troponin, aspartate aminotransferase, alanine aminotransferase, urea, and lactate, as well as proteinuria. Finally, administration of BI113823 improved the 7-day survival rate following CLP in rats. CONCLUSIONS: Administration of BI113823 reduced systemic and tissue inflammatory responses, prevented hemodynamic derangement, attenuated multiorgan injury, and improved overall survival.


Asunto(s)
Antiinflamatorios/uso terapéutico , Antagonistas del Receptor de Bradiquinina B1/uso terapéutico , Inflamación/patología , Inflamación/prevención & control , Sepsis/tratamiento farmacológico , Sepsis/patología , Animales , Modelos Animales de Enfermedad , Masculino , Ratas Wistar , Análisis de Supervivencia , Resultado del Tratamiento
6.
Toxicol Appl Pharmacol ; 305: 153-160, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27288733

RESUMEN

INTRODUCTION: The aim of the present study was to evaluate the effects of the novel kinin B1 receptor antagonist BI113823 on postinfarction cardiac remodeling and heart failure, and to determine whether B1 receptor blockade alters the cardiovascular effects of an angiotensin 1 converting enzyme (ACE) inhibitor in rats. METHODS AND RESULTS: Sprague Dawley rats were subjected to permanent occlusion of the left coronary artery. Cardiovascular function was determined at 6weeks postinfarction. Treatment with either B1 receptor antagonist (BI113823) or an ACE inhibitor (lisinopril) alone or in combination significantly reduced the heart weight-to-body weight and lung weight-to-body weight ratios, and improved postinfarction cardiac function as evidenced by greater cardiac output, the maximum rate of left ventricular pressure rise (±dP/dtmax), left ventricle ejection fraction, fractional shorting, better wall motion, and attenuation of elevated left ventricular end diastolic pressure (LVEDP). Furthermore, all three treatment groups exhibited significant reduction in cardiac interstitial fibrosis, collagen deposition, CD68 positive macrophages, neutrophils, and proinflammatory cytokine production (TNF-α and IL-1ß), compared to vehicle controls. CONCLUSION: The present study shows that treatment with the novel kinin B1 receptor antagonist, BI113823, reduces postinfarction cardiac remodeling and heart failure, and does not influence the cardiovascular effects of the ACE inhibitor.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Antagonistas del Receptor de Bradiquinina B1/uso terapéutico , Insuficiencia Cardíaca/tratamiento farmacológico , Lisinopril/uso terapéutico , Infarto del Miocardio/tratamiento farmacológico , Remodelación Ventricular/efectos de los fármacos , Animales , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Masculino , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Ratas Sprague-Dawley , Receptor de Bradiquinina B1/genética
7.
Pharmacol Res ; 104: 132-9, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26747401

RESUMEN

Kinin B1 receptors are implicated in asthmatic airway inflammation. Here we tested this hypothesis by examining the anti-inflammatory effects of BI113823, a novel non-peptide orally active kinin B1 receptor antagonist in mice sensitized to ovalbumin (OVA). Male Balb-c mice were randomly assigned to four study groups: (1) control, (2) OVA+vehicle, (3) OVA+BI113823, (4) OVA+dexamethasone. Mice were sensitized intraperitoneally with 75µg ovalbumin on days 1 and 8. On days 15-17, mice were challenged intranasally with 50µg of ovalbumin. Mice received vehicle, BI113823, or dexamethasone (positive control) on days 16-18. On day 19, bronchoalveolar lavage (BAL) and lung tissue were collected for biochemical and immuno-histological analysis. Compared to controls treatment with BI113823 significantly reduced the numbers of BAL eosinophils, macrophages, neutrophils and lymphocytes by 58.3%, 61.1%, 66.4% and 56.0%, respectively. Mice treated with dexamethasone showed similar reductions in BAL cells. Treatment with BI113823 and dexamethasone also significantly reduced total protein content, IgE, TNF-α and IL-1ß in lavage fluid, reduced myeloperoxidase activity, mucus secretion in lung tissues, and reduced the expression of B1 receptors, matrix metalloproteinase (MMP)-2 and cyclooxygenase (COX)-2 compared to vehicle-treated mice. Only BI113823 reduced MMP-9 and inducible nitric oxide synthase (iNOS). BI113823 effectively reduced OVA-induced inflammatory cell, mediator and signaling pathways equal to or greater than that seen with steroids in a mouse asthma model. BI113823 might be useful in modulating inflammation in asthma.


Asunto(s)
Antiinflamatorios/uso terapéutico , Asma/tratamiento farmacológico , Antagonistas del Receptor de Bradiquinina B1/uso terapéutico , Alérgenos , Animales , Antiinflamatorios/farmacología , Asma/inmunología , Asma/metabolismo , Asma/patología , Antagonistas del Receptor de Bradiquinina B1/farmacología , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Recuento de Células , Ciclooxigenasa 2/inmunología , Dexametasona/farmacología , Inmunoglobulina E/inmunología , Interleucina-1beta/inmunología , Pulmón/metabolismo , Pulmón/patología , Masculino , Metaloproteinasa 2 de la Matriz/inmunología , Ratones Endogámicos BALB C , Moco/metabolismo , Ovalbúmina , Factor de Necrosis Tumoral alfa/inmunología
8.
Crit Care Med ; 43(11): e499-507, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26468713

RESUMEN

OBJECTIVES: This study was undertaken to examine the effects of BI113823, a potent small molecule orally active nonpeptide B1 receptor antagonist, in an experimental model of endotoxin-induced direct lung injury in mice and indirect lung injury and survival in cecal ligation and puncture-induced polymicrobial sepsis in rats. DESIGN: Experimental, prospective study. SETTING: University research laboratory. SUBJECTS: Male BALB/c mice and male Wistar rats. INTERVENTIONS: Series 1: acute lung injury was induced in mice by intratracheal injection of lipopolysaccharide. Mice were then randomly assigned to receive treatment of vehicle, BI113823, or dexamethasone. Bronchoalveolar lavage fluid and lung tissues were analyzed for inflammatory cell influx and various histologic variables. Series 2: sepsis was induced by cecal ligation and puncture in anesthetized rats. Animals were then randomly assigned to receive treatment of vehicle or BI113823. Experiments were terminated at 20 hours and 7 days following cecal ligation and puncture, respectively. MEASUREMENTS AND MAIN RESULTS: Series 1: treatment with BI113823 significantly reduced lipopolysaccharide-induced neutrophil influx in bronchoalveolar lavage fluid. The BI113823 group had significantly lower lung vascular permeability, lung water content, myeloperoxidase activity, lung apoptosis and lung injury scores, total protein content, and tumor necrosis factor-α and interleukin-1ß levels compared with vehicle controls. In addition, nuclear factor-κB phosphorylation, nuclear translocation, and cyclooxygenase-2 and inducible nitric oxide synthase expression in the lung were attenuated in BI113823-treated animals compared with vehicle controls. Series 2: BI113823 significantly reduced sepsis-induced macrophage recruitment, protein content, and tumor necrosis factor-α and interleukin-1ß levels in lavage fluid and also reduced lung water content and plasma levels of tumor necrosis factor-α and interleukin-6 compared with vehicle controls. Most importantly, treatment with BI113823 significantly improved survival following severe sepsis in rats. CONCLUSIONS: Administration of B1 receptor antagonist BI113823 significantly reduced endotoxin-induced direct lung injury and also reduced sepsis-induced lung inflammatory response. Most importantly, BI113823 improved survival following severe polymicrobial sepsis.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Bradiquinina/antagonistas & inhibidores , Citocinas/metabolismo , Dexametasona/uso terapéutico , Macrófagos/efectos de los fármacos , Lesión Pulmonar Aguda/mortalidad , Lesión Pulmonar Aguda/fisiopatología , Animales , Western Blotting , Bradiquinina/administración & dosificación , Líquido del Lavado Bronquioalveolar/citología , Ciclooxigenasa 2/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Inmunohistoquímica , Mediadores de Inflamación/metabolismo , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/metabolismo , Macrófagos/citología , Masculino , Ratones , Ratones Endogámicos BALB C , Distribución Aleatoria , Ratas , Ratas Wistar , Valores de Referencia , Tasa de Supervivencia , Resultado del Tratamiento
9.
Pharmacol Res ; 90: 18-24, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25258294

RESUMEN

This study examined responses of isolated pig coronary arteries after kinin B1 receptor induction by endotoxin. Des-Arg9-bradykinin (DBK) induced concentration-dependent, endothelium-independent contractions in lipopolysaccharide (LPS)-treated but not untreated arterial rings. The B1-receptor antagonist SSR240612, but not the B2-receptor antagonist HOE140, prevented the endothelium-independent contractions to DBK. The DBK-induced contractions were blocked by indomethacin (nonselective cyclooxygenase [COX] inhibitor), celecoxib (selective COX-2 inhibitor), and terbogrel (thromboxane-prostanoid [TP] receptor antagonist) but not valeryl salicylate (selective COX-1 inhibitor), AH6809 (an E prostanoid [EP] and PGD2 receptor [DP1] receptor antagonist), AL 8810 (a selective PGF2α [FP] receptor antagonist), or RO1138452 (a selective I prostanoid [IP] receptor antagonist). They were attenuated by N-(p-amylcinnamoyl) anthranilic acid (ACA), and by DETCA plus tiron but not by l-NAME. Quantitative RT-PCR revealed excessive up-regulations of mRNA expressions of B1 receptors, COX-2, and thromboxane A synthase 1 (TBXAS1) following LPS incubation, but not of B2 receptors or COX-1. The present data demonstrate that B1 receptors are coupled to COX-2 in causing endothelium-independent contractions in endotoxin-treated pig coronary arteries. Accordingly, kinin B1 receptor induction during inflammation may have a pathological significance in the vasculature, particular in coronary arteries with dysfunctional endothelial cells.


Asunto(s)
Vasos Coronarios/fisiología , Ciclooxigenasa 2/fisiología , Receptor de Bradiquinina B1/fisiología , Vasoconstricción/fisiología , Animales , Bradiquinina/análogos & derivados , Bradiquinina/farmacología , Antagonistas del Receptor de Bradiquinina B1/farmacología , Vasos Coronarios/efectos de los fármacos , Ciclooxigenasa 1/genética , Ciclooxigenasa 2/genética , Dioxoles/farmacología , Endotelio Vascular , Técnicas In Vitro , Lipopolisacáridos/farmacología , ARN Mensajero/biosíntesis , Receptor de Bradiquinina B1/genética , Receptor de Bradiquinina B2/genética , Sulfonamidas/farmacología , Porcinos , Tromboxano-A Sintasa/genética , Vasoconstricción/efectos de los fármacos
10.
J Cardiovasc Pharmacol ; 64(3): 209-17, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25192543

RESUMEN

This study examined the vascular effects of bradykinin in health and vascular inflammation comparing responses of isolated pig coronary arteries in the absence and presence of endotoxins. Bradykinin induced contractions in lipopolysaccharide-treated, but not untreated, arterial rings without endothelium. The B2-receptor antagonist HOE140, but not the B1-receptor inhibitor SSR240612, blocked these endothelium-independent contractions in response to bradykinin. The bradykinin-induced contractions were blocked by indomethacin, celecoxib, and terbogrel but not valeryl salicylate, AH6809, AL 8810, or RO1138452. They were attenuated by N-(p-amylcinnamoyl) anthranilic acid, and by diethyldithiocarbamate plus tiron but not by L-NAME. Quantitative reverse-transcription polymerase chain reaction revealed significant upregulations of messenger RNA expressions of B1 receptors, COX-2, and thromboxane A synthase 1 (TBXAS1) following lipopolysaccharide incubation but not of B2 receptors or COX-1. The present data demonstrate that bradykinin induces contractions mediated by the COX-2 pathway in endotoxin-treated pig coronary arteries. These results support differential roles of bradykinin in health and disease.


Asunto(s)
Bradiquinina/metabolismo , Vasos Coronarios/metabolismo , Ciclooxigenasa 2/metabolismo , Inflamación/patología , Animales , Bradiquinina/farmacología , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/patología , Ciclooxigenasa 2/genética , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Endotoxinas/farmacología , Lipopolisacáridos/farmacología , Contracción Muscular/efectos de los fármacos , ARN Mensajero , Receptor de Bradiquinina B1/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Porcinos , Tromboxano-A Sintasa/genética , Regulación hacia Arriba
11.
Eur J Med Res ; 28(1): 71, 2023 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-36755351

RESUMEN

BACKGROUND: Airway remodeling is an important pathological feature of chronic airway diseases, which leads to a progressive decline in lung function. The present study examined the anti-remodeling and anti- inflammatory effect of BIBF1000, a triple-tyrosine kinase inhibitor that targets VEGF, PDGF, and FGF receptor signaling in a mouse model of repeated ovalbumin (OVA) challenges. METHODS: Female Balb-c mice were immunized intraperitoneally on days 0 and 12 with 50 µg ovalbumin plus 1 mg of Al(OH)3 in 200 µl saline. Intranasal OVA challenges (20 µg/50 µl in PBS) were administered on days 26, 29, and 31, and were repeated twice a week for 3 months. Animals received vehicle or BIBF1000 (25 mg/kg, b.i.d.) through gavage from day 26 to the end of fourth month. On day 120, bronchoalveolar lavage (BAL) and lung tissue were collected for biochemical and immunohistological analysis. RESULTS: Compared to vehicle controls, treatment with BIBF1000 reduced the numbers of BAL eosinophils, macrophages, neutrophils, and lymphocytes by 70.0%, 57.9%, 47.5%, and 63.0%, respectively, and reduced IL-5 and IL-13 in BAL. Treatment with BIBF1000 reduced airway mucus secretion, peribronchial fibrosis, small airway, and pulmonary arterial wall thickness, compared to vehicle controls. Furthermore, treatment with BIBF1000 also reduced the expression of inflammatory mediators (TNF-α, IL-1ß, IL-5, IL-13, MMP-2, MMP-9, COX-2, and iNOS) and inhibited ERK and AKT phosphorylation. CONCLUSIONS: The protective effect afforded by triple-tyrosine kinase inhibition with BIBF1000 in reducing allergen-induced airway and arterial remodeling was associated with down-regulation of inflammatory mediators, as well as inhibition of ERK and AKT signaling pathways.


Asunto(s)
Alérgenos , Interleucina-13 , Remodelación Vascular , Animales , Femenino , Ratones , Alérgenos/farmacología , Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Interleucina-13/metabolismo , Interleucina-5/metabolismo , Pulmón/patología , Ratones Endogámicos BALB C , Ovalbúmina , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , /farmacología
12.
Clin Exp Pharmacol Physiol ; 37(3): 337-42, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19769605

RESUMEN

1. The aim of the present study was to examine the effects of inhibition of the Na(+)/H(+) exchanger (NHE-1) on cardiopulmonary performance, oxygen carrying capacity and tissue inflammation in a pig model of traumatic haemorrhage-resuscitation. 2. In 12 instrumented anaesthetized pigs, traumatic haemorrhage was modelled by producing tibia fractures, followed by haemorrhage of 25 mL/kg for 20 min, and then a 4 mm hepatic arterial tear with surgical repair after 20 min. Animals then underwent low-volume fluid resuscitation with either Hextend (vehicle; n = 6; Hospira, Lake Forest, IL, USA) or 3 mg/kg BIIB513 (an NHE-1 inhibitor) + Hextend (n = 6). The experiment was terminated 6 h after the beginning of resuscitation. 3. Compared with vehicle-treated controls, the addition of NHE-1 inhibition with BIIB513 significantly improved the left ventricle stroke work index and attenuated increases in pulmonary arterial pressure and pulmonary vascular resistance. Furthermore, BIIB513 treatment significantly increased the oxygenated haemoglobin ratio, blood oxygen content and mixed venous blood oxygen saturation and improved blood oxygen delivery. In addition, BIIB513 treatment reduced lung tissue levels of interleukin-6 by 80%, tumour necrosis factor-alpha by 37% and myeloperoxidase activity by 38%. Nuclear factor-kappaB DNA binding activity in the lung was also slightly and significantly attenuated following BIIB513 treatment. 4. In conclusion, the present study shows that NHE-1 inhibition facilitates the response to fluid resuscitation after traumatic haemorrhage by improving cardiac function, pulmonary vascular function and oxygen carrying capacity, which results in reduced tissue inflammatory injury.


Asunto(s)
Reanimación Cardiopulmonar/métodos , Mediadores de Inflamación/fisiología , Oxígeno/metabolismo , Choque Hemorrágico/metabolismo , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Intercambiadores de Sodio-Hidrógeno/metabolismo , Animales , Mediadores de Inflamación/farmacología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Mesilatos/farmacología , Mesilatos/uso terapéutico , Miocardio/metabolismo , Miocardio/patología , Resucitación/métodos , Choque Hemorrágico/tratamiento farmacológico , Choque Hemorrágico/patología , Porcinos
13.
Prostaglandins Other Lipid Mediat ; 90(1-2): 21-5, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19559811

RESUMEN

The aim of the present study was to compare the effects of genetic mPGES-1 loss and COX-2 inhibition on myocardial damage after coronary occlusion. mPGES-1(-/-) mice and their wild-type littermates were injected with vehicle or COX-2 inhibitor (celecoxib), and 30min later the left coronary artery was surgically occluded. At 24h, myocardial infarct (MI) volume was measured histologically. Post-MI survival was reduced in WT mice receiving celecoxib (12/20) compared with vehicle-treated controls (12/12) or the loss of mPGES-1 (13/13) together with increased phosphokinase (CPK) and cardiac troponin-I release. Endogenous mPGES-1 expression was unchanged by ischemia in WT mice and absent in mPGES-1(-/-) hearts. COX-2 expression was markedly increased at 24h after MI in WT hearts; this upregulation was largely attenuated in mPGES-1(-/-) mice. We conclude that loss of mPGES-1 prevents the upregulation of COX-2 after myocardial infarct, and in contrast to inhibition of COX-2, does not increase ischemic myocardial damage.


Asunto(s)
Inhibidores de la Ciclooxigenasa 2/farmacología , Eliminación de Gen , Oxidorreductasas Intramoleculares/deficiencia , Oxidorreductasas Intramoleculares/genética , Microsomas/enzimología , Isquemia Miocárdica/enzimología , Isquemia Miocárdica/genética , Animales , Oclusión Coronaria/complicaciones , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Regulación Enzimológica de la Expresión Génica , Ratones , Infarto del Miocardio/complicaciones , Infarto del Miocardio/patología , Isquemia Miocárdica/complicaciones , Isquemia Miocárdica/patología , Prostaglandina-E Sintasas , ARN Mensajero/genética , ARN Mensajero/metabolismo
14.
Transl Res ; 203: 15-30, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30142307

RESUMEN

The present study examined the effects of simultaneous inhibition of vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF) and fibroblast growth factor (FGF) receptor signaling with BIBF1000, a novel triple tyrosine kinase inhibitor on preventing and reversing the progression of severe pulmonary arterial hypertension (PAH) in an experimental model in rats. Left pneumonectomized male Wistar rats were injected with monocrotaline to induce PAH. Treatment with BIBF1000 from day 1 to day 21 after monocrotaline injection attenuated PAH development, as evidenced by lower values for pulmonary artery pressure (mPAP), right ventricular pressure (RVSP), pulmonary arterial neointimal formation, and the ratio of right ventricular weight to left ventricular and septum weight [RV/(LV+S)] on day 21 compared to control rats. Treatment with BIBF1000 from day 21 to day 42 after monocrotaline injection reversed established PAH as shown by normalized values for mPAP and RVSP, RV/(LV+S) ratio, pulmonary arterial occlusion scores, levels of heart and lung fibrosis, as well as improved survival. Treatment with BIBF1000 reduced inflammatory cell recruitment in bronchoalveolar lavage and lung tissues, reduced CD-68 positive macrophages and expression of proliferating cell nuclear antigen in the perivascular areas, and reduced TNF-α and growth factor productions, and inhibited the phosphorylation of AKT and GSK3ß in lungs. In addition, BIBF1000 inhibited pulmonary artery smooth muscle cells migration and proliferation from rat pulmonary artery explant cultures. Simultaneous inhibition of VEGF, PDGF, and FGF receptor signaling by BIBF1000 prevents and reverses the progression of severe pulmonary arterial hypertension and vascular remodeling in this experimental model.


Asunto(s)
Hipertensión Pulmonar/tratamiento farmacológico , Indoles/farmacología , Neointima , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Inflamación , Masculino , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular
15.
Trends Pharmacol Sci ; 28(11): 580-7, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17963849

RESUMEN

Migraine is a complex, debilitating neurovascular disorder. Although knowledge on the main molecular players is still incomplete, recent preclinical and clinical findings indicate that there is a clear correlation between migraine-associated headache and the release of the neuropeptide calcitonin gene-related peptide (CGRP). BIBN4096 was the first CGRP antagonist to be tested in clinical trials for the treatment of migraine. The proven efficacy of this agent, and also the CGRP antagonist MK-0974, to alleviate acute migraine headache provided significant support for the hypothesis that CGRP has an important role in migraine pathophysiology. Moreover, the recently published results from Phase II trials are encouraging and suggest that this new type of drug might offer advantages over existing therapies for patients suffering from migraine and related headaches.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/antagonistas & inhibidores , Trastornos Migrañosos/tratamiento farmacológico , Animales , Azepinas/química , Azepinas/farmacología , Azepinas/uso terapéutico , Péptido Relacionado con Gen de Calcitonina/fisiología , Ensayos Clínicos como Asunto , Dipéptidos/química , Dipéptidos/farmacología , Dipéptidos/uso terapéutico , Humanos , Imidazoles/química , Imidazoles/farmacología , Imidazoles/uso terapéutico , Trastornos Migrañosos/fisiopatología , Modelos Biológicos , Estructura Molecular , Quinazolinas/química , Quinazolinas/farmacología , Quinazolinas/uso terapéutico
16.
Shock ; 29(4): 519-25, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17724433

RESUMEN

Severe blood loss is a major cause of death occurring within hours of traumatic injury. Na+/H+ exchange (NHE-1) activity is an important determinant of the extent of ischemic myocardial injury. The goal of the present study was to test the hypothesis that NHE-1 inhibition delays the onset of hypovolemic circulatory shock, thereby preventing early death due to severe hemorrhage in pigs. Severe hypovolemia was studied in 16 (25.2 kg) anesthetized male pigs in steps of 10-, 20-, 30-, 40-, and 50-mL kg(-1) blood loss, each in 30-min intervals. Shed blood resuscitation was started 30 min after 50 mL kg(-1) blood loss. The experiment was terminated after 3 h of resuscitation. Eight pigs were used as seline control. Eight pigs received 3 mg kg(-1) benzamide, N-(aminoiminomethyl)-4-[4-(2-furanylcarbonyl)-1-piperazinyl]-3-(methylsulfonyl), methanesulfonate (NHE-1 inhibitor) 15 min before hemorrhage. Seven control pigs died at 40- to 50-mL kg(-1) blood loss. One control pig survived initial resuscitation but died soon after. In contrast, all animals treated with NHE-1 inhibitor survived the entire protocol. In control animals, cardiac output and MAP gradually decreased at each step of blood loss with marked increase in heart rate. Cardiovascular decompensation occurred at 40 mL kg(-1) blood loss. Na+/H+ exchange inhibition increased oxygen delivery, attenuated cardiovascular decompensation, delayed the onset of irreversible hypovolemic circulatory shock, and enabled resuscitation to survival. Echocardiography analysis showed that myocardial hypercontracture gradually developed with each step of blood loss in control animals, but this hypercontracture was attenuated in the animals receiving the NHE-1 inhibitor. We conclude that NHE-1 inhibition attenuates ischemic myocardial hypercontracture, cardiovascular decompensation, delays the onset of hypovolemic circulatory shock, and prevents early death in severe hemorrhage.


Asunto(s)
Sistema Cardiovascular/fisiopatología , Choque/fisiopatología , Intercambiadores de Sodio-Hidrógeno/fisiología , Animales , Benzamidas/farmacología , Gasto Cardíaco/efectos de los fármacos , Sistema Cardiovascular/efectos de los fármacos , Sistema Cardiovascular/patología , Ecocardiografía , Hemodinámica/efectos de los fármacos , Masculino , Mesilatos/farmacología , Daño por Reperfusión Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/prevención & control , Choque/prevención & control , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Porcinos
17.
Eur J Pharmacol ; 580(1-2): 55-62, 2008 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-18031735

RESUMEN

Small molecules interacting with aminergic G-protein coupled receptors represent a number of very successful drugs. G-protein coupled receptors continue to be a significant group of targets for pharmaceutical intervention, and modifying their activity through small molecules is a major focus of drug development. Previously, these small molecules could be easily fit in models, as agonists, partial agonists or antagonists. More recently, however, these lines have been blurred as it is increasingly recognized that ligands can interact with receptors in various ways. Analysis of beta-adrenoceptors has revealed that several sites or states exist for the individual receptors. The putative atypical beta(4)-adrenoceptor identified on heart and adipose tissue is now recognized as a unique beta(1)-adrenoceptor state. Similarly, a unique beta(3)-adrenoceptor state has been identified using the aryloxypropanolamine CGP-12,177 and cloned receptor systems. Here we expand upon these observations, by describing an atypical state of the beta(3)-adrenoceptor that exists endogenously in adipose tissue. Furthermore, we describe novel arylethanolamine ligands that interact with this atypical state of the beta(3)-adrenoceptor with high affinity and provide additional tools to investigate the atypical beta(3)-adrenoceptor state to determine whether it can be influenced for therapeutic purposes.


Asunto(s)
Adipocitos Blancos/metabolismo , Lipólisis/fisiología , Receptores Adrenérgicos beta 3/metabolismo , Agonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/farmacología , Animales , AMP Cíclico/metabolismo , Etanolaminas/farmacología , Femenino , Ligandos , Lipólisis/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Ratas , Ratas Wistar , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 3/genética
18.
Behav Brain Res ; 350: 129-138, 2018 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-29738803

RESUMEN

BACKGROUND: Burrowing is a rodent behavior validated as a robust and reproducible outcome measure to infer the global effect of pain in several inflammatory pain models. However, less is known about the effect of analgesics on burrowing in neuropathic pain models and no studies have determined burrowing performance in models of diabetes-associated neuropathic pain. OBJECTIVE: To compare the sensitivity of the burrowing assay in different neuropathic pain models: mononeuropathic pain and diabetic polyneuropathy. METHODS: Burrowing performance was determined by the amount of substrate left in a hollow tube by rats with chronic constriction injury (CCI). In addition, burrowing performance, locomotion and pain development was assessed in the Zucker diabetic fatty (ZDF) rat model, resembling type-2 diabetes. Efficacy of clinically-active reference drugs (opioids, gabapentin and/or pregabalin) were investigated in these models. Burrowing behavior was additionally assessed in a second model, induced by streptozotocin (STZ) treatment, resembling type-1 diabetes. RESULTS: In the CCI model, moderate but consistent burrowing deficits were observed that persisted over a period of ≥20 days. Systemic administration of morphine, pregabalin and gabapentin reversed this deficit. In contrast, none of the reference drugs improved marked burrowing deficits detected in ZDF rats, and pregabalin did not reverse severe burrowing deficits observed in STZ rats. CONCLUSIONS: Burrowing performance cannot necessarily be used as pain-related readout across pain models and largely depends on the model used, at least in models of neuropathy. Specifically, analgesic drug effects might be masked by general diabetes-associated alteration of the animals' well-being, resulting in false negative outcomes.


Asunto(s)
Conducta Animal , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 1/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Actividad Motora , Neuralgia/fisiopatología , Analgésicos/farmacología , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Masculino , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Neuralgia/tratamiento farmacológico , Neuralgia/etiología , Distribución Aleatoria , Ratas Wistar , Ratas Zucker
19.
J Cardiovasc Pharmacol Ther ; 12(1): 61-8, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17495259

RESUMEN

Monocyte chemoattractant protein-1 (MCP-1) is a stimulator of collateral artery growth and has been shown to increase collateral artery conductance in rabbits and pigs. The minimal infusion duration and the minimally effective dose of MCP-1 are currently unknown, as is the sustainability of the therapeutic effect over a longer observation period than tested before. MCP-1 was infused intra-arterially in pigs after unilateral femoral artery occlusion in different doses and infusion durations between 2 hours and 2 weeks. Two weeks after ligation, arterial conductance under maximal vasodilatation was measured. The long-term efficacy was investigated in 2 additional groups of animals after 6 weeks. Infusion with 2 microg/min of MCP-1 for 6 hours was sufficient to double arterial conductance, and arterial conductance after 6 weeks was still significantly increased.


Asunto(s)
Arterias/efectos de los fármacos , Quimiocina CCL2/farmacología , Enfermedades Vasculares Periféricas/tratamiento farmacológico , Animales , Arterias/crecimiento & desarrollo , Arterias/fisiopatología , Células CHO , Quimiocina CCL2/administración & dosificación , Quimiocina CCL2/genética , Circulación Colateral/efectos de los fármacos , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Arteria Femoral/cirugía , Humanos , Infusiones Intraarteriales , Ligadura/efectos adversos , Masculino , Enfermedades Vasculares Periféricas/etiología , Enfermedades Vasculares Periféricas/fisiopatología , Receptores CCR2 , Receptores de Quimiocina/genética , Receptores de Quimiocina/fisiología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Porcinos , Porcinos Enanos , Factores de Tiempo , Resultado del Tratamiento
20.
Neurosci Lett ; 619: 162-7, 2016 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-26987721

RESUMEN

The current study assessed whether antidepressant and/or antinociceptive drugs, duloxetine, fluoxetine as well as (±)-8-hydroxy-2-[di-n-propylamino] tetralin (8-OH-DPAT), are able to reverse depression-like behaviour in animals with chronic neuropathic pain. Chronic constriction injury (CCI) of the sciatic nerve in rats was selected as neuropathic pain model. Mechanical hypersensitivity and depression-like behaviour were evaluated 4 weeks after surgery by "electronic algometer" and forced swimming test (FST), which measured the time of immobility, and active behaviours climbing and swimming. The selective noradrenergic and serotonergic uptake blocker duloxetine (20mg/kg) and the selective 5-HT1A agonist 8-OH-DPAT (0.5mg/kg) significantly reversed both mechanical hypersensitivity and depression-like behaviour in CCI animals. Duloxetine significantly reversed depression-like behaviour in CCI rats by increasing the time of climbing and swimming, while 8-OH-DPAT attenuated depression-like behaviour mainly by increasing the time of swimming. However, the selective serotonergic uptake blocker fluoxetine (20mg/kg) failed to attenuate mechanical hypersensitivity and depression-like behaviour, possibly due to confounding pro-nociceptive actions at 5-HT3 receptors. These data suggest to target noradrenergic and 5-HT1A receptors for treatment of chronic pain and its comorbidity depression.


Asunto(s)
8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Analgésicos/farmacología , Antidepresivos/farmacología , Depresión/tratamiento farmacológico , Clorhidrato de Duloxetina/farmacología , Fluoxetina/farmacología , Neuralgia/tratamiento farmacológico , 8-Hidroxi-2-(di-n-propilamino)tetralin/uso terapéutico , Neuronas Adrenérgicas/metabolismo , Analgésicos/uso terapéutico , Animales , Antidepresivos/uso terapéutico , Conducta Animal/efectos de los fármacos , Enfermedad Crónica , Depresión/metabolismo , Depresión/fisiopatología , Depresión/psicología , Clorhidrato de Duloxetina/uso terapéutico , Fluoxetina/uso terapéutico , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/fisiopatología , Masculino , Neuralgia/metabolismo , Neuralgia/fisiopatología , Neuralgia/psicología , Norepinefrina/metabolismo , Ratas Wistar , Neuronas Serotoninérgicas/metabolismo , Serotonina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA