Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biol Chem ; 397(12): 1217-1222, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27622831

RESUMEN

Genetic and pharmacological studies, clinical and experimental, focused on kallikrein-K1, kinin receptors and ACE/kininase II suggest that kinin release in the settings of ischemia or diabetes reduces organ damage, especially in the heart and kidney. Kinin bioavailability may be a limiting factor for efficacy of current kinin-potentiating drugs, like ACE inhibitors. Primary activation of kinin receptors by prototypic pharmacological agonists, peptidase-resistant, selective B1 or B2, displays therapeutic efficacy in experimental cardiac and peripheral ischemic and diabetic diseases. B1R agonism was especially efficient in diabetic animals and had no unwanted effects. Clinical development of kinin receptor agonists may be warranted.


Asunto(s)
Diabetes Mellitus/metabolismo , Diabetes Mellitus/terapia , Isquemia/metabolismo , Isquemia/terapia , Sistema Calicreína-Quinina , Animales , Diabetes Mellitus/tratamiento farmacológico , Humanos , Isquemia/tratamiento farmacológico , Sistema Calicreína-Quinina/efectos de los fármacos
2.
J Cardiovasc Pharmacol ; 63(3): 274-81, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24220315

RESUMEN

Coronary endothelial dysfunction is involved in cardiac ischemia-reperfusion (IR) injury. Vascular endothelial growth factor (VEGF) activates endothelial cells and exerts cardioprotective effects in isolated hearts. The recently discovered viper venom protein called increasing capillary permeability protein (ICPP) exerts VEGF-like effects in endothelial cells. We examined whether VEGF or ICPP can influence IR outcome in vivo in mice. Dosages of VEGF and ICPP were determined by preliminary blood pressure study. In IR, both the proteins administered intravenously at reperfusion reduced infarct size (IS) by 57% for VEGF and 52% for ICPP (P < 0.01). Pretreatment with a selective VEGFR2 receptor antagonist abolished the reduction in IS. VEGF and ICPP induced ERK phosphorylation in the myocardium. IR triggered mitochondrial pore opening and impaired mitochondrial respiratory function. These effects of IR were prevented by VEGF or ICPP, which increased mitochondrial calcium retention capacity by 37% compared with saline (P < 0.05) and improved mitochondrial respiratory function (by 71% and 65%, respectively for state 3, and 51% and 38% for state 4, P < 0.01 for VEGF). Thus, intravenous administration of VEGF or ICPP at reperfusion largely reduces IS in IR, through stimulation of VEGFR2 receptors. This effect is mediated, at least in part, by improvement of IR-induced mitochondrial dysfunction.


Asunto(s)
Daño por Reperfusión Miocárdica/tratamiento farmacológico , Proteínas/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología , Venenos de Víboras/química , Animales , Calcio/metabolismo , Cardiotónicos/administración & dosificación , Cardiotónicos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/patología , Infarto del Miocardio/patología , Infarto del Miocardio/prevención & control , Isquemia Miocárdica/tratamiento farmacológico , Isquemia Miocárdica/patología , Daño por Reperfusión Miocárdica/patología , Proteínas/administración & dosificación , Proteínas/química , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
3.
J Pharmacol Exp Ther ; 346(1): 23-30, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23591995

RESUMEN

Cardiac ischemia is a leading cause of death, especially in diabetic patients. The diabetic ischemic heart is resistant experimentally to established cardioprotective treatments. New pharmacological approaches to cardiac protection are warranted. The kallikrein-kinin system is involved in myocardial protection in ischemia. Respective roles of B1 (B1R) and B2 (B2R) receptors remain controversial. We tested whether pharmacological activation of kinin receptors may have therapeutic effect in cardiac ischemia-reperfusion in nondiabetic (NDiab) and diabetic (Diab) mice. We assessed effect on infarct size (IS) and signaling pathways involved in myocardial protection of potent selective pharmacological agonists of B1R or B2R given at reperfusion. In NDiab mice, a B2R agonist reduced IS significantly by 47%, similarly to ramiprilat or ischemic postconditioning, via activation of phosphoinositide 3 kinase/Akt pathway leading to inhibition of glycogen synthase kinase-3ß (GSK-3ß). B1R agonist had no effect on IS. In contrast, in Diab mice, the B2R agonist, ramiprilat, or ischemic postconditioning failed to reduce IS but a B1R agonist significantly reduced IS by 44% via activation of phosphoinositide 3 kinase/Akt and extracellular signal-regulated kinase 1/2, both leading to GSK-3ß inhibition. Differential effect of B2R or B1R agonists in NDiab and Diab mice can be linked to inactivation of B2R signaling and induction of B1R in heart of Diab mice. Thus, a pharmacological B2R agonist is cardioprotective in acute ischemia in nondiabetic animals. B1R agonist overcomes resistance of diabetic heart to cardioprotective treatments. Pharmacological activation of B1R and B2R may become a treatment for diabetic and nondiabetic patients, respectively, in acute coronary syndromes.


Asunto(s)
Bradiquinina/análogos & derivados , Cardiotónicos/uso terapéutico , Diabetes Mellitus Tipo 1/complicaciones , Isquemia Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/prevención & control , Receptor de Bradiquinina B1/agonistas , Receptor de Bradiquinina B2/agonistas , Animales , Bradiquinina/administración & dosificación , Bradiquinina/efectos adversos , Bradiquinina/uso terapéutico , Cardiotónicos/administración & dosificación , Cardiotónicos/efectos adversos , Diabetes Mellitus Tipo 1/metabolismo , Relación Dosis-Respuesta a Droga , Resistencia a Medicamentos , Regulación de la Expresión Génica , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Isquemia Miocárdica/complicaciones , Isquemia Miocárdica/metabolismo , Miocardio/metabolismo , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Receptor de Bradiquinina B1/genética , Receptor de Bradiquinina B1/metabolismo , Receptor de Bradiquinina B2/genética , Receptor de Bradiquinina B2/metabolismo , Regulación hacia Arriba/efectos de los fármacos
4.
Biol Chem ; 394(3): 329-33, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23324381

RESUMEN

Tissue kallikrein has been suggested to be involved in blood pressure regulation and in protection against hypertension. However, this hypothesis remains debated. Recently, murine genetic models of kallikrein deficiency have been engineered and partial genetic deficiency in kallikrein activity has been characterized in humans. Studies in kallikrein-deficient mice indicate that kallikrein indeed influences blood pressure in the setting of mineralocorticoid excess and salt retention but not in normotensive animals and in high renin hypertension. These observations suggest that kallikrein can have antihypertensive function in physiological situations where sodium retention can trigger blood pressure elevation.


Asunto(s)
Presión Sanguínea/fisiología , Hipertensión/fisiopatología , Calicreínas de Tejido/deficiencia , Calicreínas de Tejido/metabolismo , Animales , Humanos , Ratones , Ratones Transgénicos , Calicreínas de Tejido/genética
5.
Circ Res ; 108(3): 284-93, 2011 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-21164105

RESUMEN

RATIONALE: Homing of proangiogenic cells (PACs) is guided by chemoattractants and requires proteases to disrupt the extracellular matrix. The possibility that PAC recruitment involves an interaction between proteases and chemotactic factor receptors remains largely unexplored. OBJECTIVE: To determine the role of human tissue kallikrein (hK1) in PAC invasion and its dependency on kinin receptor signaling. METHODS AND RESULTS: Human mononuclear cells (MNCs) and culture-selected PACs express and release mature hK1 protein. HK1 gene (KLK1) silencing reduced PACs migratory, invasive, and proangiogenic activities. KLK1-knockout mouse bone marrow-derived MNCs showed similar impairments and were unable to support reparative angiogenesis in a mouse model of peripheral ischemia. Conversely, adenovirus-mediated KLK1 (Ad.KLK1) gene transfer enhanced PAC-associated functions, whereas the catalytically inactive variant R53H-KLK1 was ineffective. HK1-induced effects are mediated by a kinin B(2) receptor (B(2)R)-dependent mechanism involving inducible nitric oxide synthase and metalloproteinase-2 (MMP2). Lower hK1 protein levels were observed in PACs from type 2 diabetic (T2D) patients, whereas KLK1 mRNA levels were similar to those of healthy subjects, suggesting a post-transcriptional defect. Furthermore, B(2)R is normally expressed on T2D-PACs but remains uncoupled from downstream signaling. Importantly, whereas Ad.KLK1 alone could not restore T2D-PAC invasion capacity, combined KLK1 and B(2)R expression rescued the diabetic phenotype. CONCLUSIONS: This study reveals new interactive components of the PACs invasive machinery, acting via protease- and kinin receptor-dependent mechanisms.


Asunto(s)
Movimiento Celular/fisiología , Calicreínas/metabolismo , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Neovascularización Fisiológica/fisiología , Adulto , Anciano , Animales , Estudios de Casos y Controles , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Regulación hacia Abajo , Femenino , Miembro Posterior/irrigación sanguínea , Humanos , Isquemia/metabolismo , Calicreínas/genética , Cininas/metabolismo , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Modelos Animales , Óxido Nítrico Sintasa/metabolismo , ARN Mensajero/metabolismo , Receptor de Bradiquinina B2/metabolismo , Transducción de Señal/fisiología
6.
FASEB J ; 24(12): 4691-700, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20667972

RESUMEN

Angiotensin I-converting enzyme (ACE; kininase II) levels in humans are genetically determined. ACE levels have been linked to risk of myocardial infarction, but the association has been inconsistent, and the causality underlying it remains undocumented. We tested the hypothesis that genetic variation in ACE levels influences myocardial tolerance to ischemia. We studied ischemia-reperfusion injury in mice bearing 1 (ACE1c), 2 (ACE2c, wild type), or 3 (ACE3c) functional copies of the ACE gene and displaying an ACE level range similar to humans. Infarct size in ACE1c was 29% lower than in ACE2c (P<0.05). Pretreatment with a kinin B2 receptor antagonist suppressed this reduction. In ACE3c, infarct size was the same as in ACE2c. But ischemic preconditioning, which reduced infarct size in ACE2c (-63%, P<0.001) and ACE1c (-52%, P<0.05), was not efficient in ACE3c (-2%, NS, P<0.01 vs. ACE2c). In ACE3c, ischemic preconditioning did not decrease myocardial inflammation or cardiomyocyte apoptosis. Pretreatment with a renin inhibitor had no cardioprotective effect in ACE2c, but in ACE3c partially restored (38%) the cardioprotection of ischemic preconditioning. Thus, a modest genetic increase in ACE impairs myocardial tolerance to ischemia. ACE level plays a critical role in cardiac ischemia, through both kinin and angiotensin mediated mechanisms.


Asunto(s)
Corazón/efectos de los fármacos , Infarto del Miocardio/enzimología , Isquemia Miocárdica/enzimología , Miocardio/enzimología , Peptidil-Dipeptidasa A/metabolismo , Daño por Reperfusión/genética , Amidas/farmacología , Angiotensina I/farmacología , Angiotensina II/farmacología , Animales , Apoptosis/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Bradiquinina/análogos & derivados , Bradiquinina/farmacología , Antagonistas de los Receptores de Bradiquinina , Fumaratos/farmacología , Cininas/farmacología , Pulmón/enzimología , Ratones , Ratones Mutantes , Infarto del Miocardio/genética , Isquemia Miocárdica/genética , Peptidil-Dipeptidasa A/genética , Renina/antagonistas & inhibidores
7.
Arterioscler Thromb Vasc Biol ; 29(5): 657-64, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19164804

RESUMEN

OBJECTIVE: Human Tissue Kallikrein (hKLK1) overexpression promotes an enduring neovascularization of ischemic tissue, yet the cellular mechanisms of hKLK1-induced arteriogenesis remain unknown. Furthermore, no previous study has compared the angiogenic potency of hKLK1, with its loss of function polymorphic variant, rs5515 (R53H), which possesses reduced kinin-forming activity. METHODS AND RESULTS: Here, we demonstrate that tissue kallikrein knockout mice (KLK1-/-) show impaired muscle neovascularization in response to hindlimb ischemia. Gene-transfer of wild-type Ad.hKLK1 but not Ad.R53H-hKLK1 was able to rescue this defect. Similarly, in the rat mesenteric assay, Ad.hKLK1 induced a mature neovasculature with increased vessel diameter through kinin-B2 receptor-mediated recruitment of pericytes and vascular smooth muscle cells, whereas Ad.R53H-hKLK1 was ineffective. Moreover, hKLK1 but not R53H-hKLK1 overexpression in the zebrafish induced endothelial precursor cell migration and vascular remodeling. Furthermore, Ad.hKLK1 activates metalloproteinase (MMP) activity in normoperfused muscle and fails to promote reparative neovascularization in ischemic MMP9-/- mice, whereas its proarteriogenic action was preserved in ApoE-/- mice, an atherosclerotic model of impaired angiogenesis. CONCLUSIONS: These results demonstrate the fundamental role of endogenous Tissue Kallikrein in vascular repair and provide novel information on the cellular and molecular mechanisms responsible for the robust arterialization induced by hKLK1 overexpression.


Asunto(s)
Miembro Posterior/irrigación sanguínea , Neovascularización Fisiológica/fisiología , Circulación Esplácnica/fisiología , Calicreínas de Tejido/fisiología , Animales , Humanos , Isquemia/fisiopatología , Sistema Calicreína-Quinina/fisiología , Masculino , Metaloproteinasa 9 de la Matriz/fisiología , Ratones , Ratones Noqueados , Ratas , Cicatrización de Heridas/fisiología , Pez Cebra
8.
Eur J Heart Fail ; 10(4): 343-51, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18343196

RESUMEN

BACKGROUND: Tissue kallikrein (TK) is a major kinin-releasing enzyme present in arteries. TK is involved in cardioprotection in the setting of acute myocardial ischaemia but its role in post-ischaemic heart failure (HF), a major cause of delayed mortality after myocardial infarction (MI), is unknown. AIM: To determine whether TK deficiency in the mouse influences survival and cardiac remodelling after MI. METHODS: MI was induced in 10 week-old male TK-deficient mice and wild-type littermates. Survival was assessed up to 14 months. Cardiac morphological and functional parameters were serially measured by echocardiography. In another experiment, myocardial capillary density and NOS content were evaluated at 3 months. RESULTS: Infarct size was similar in both genotypes. MI resulted in severe cardiac dysfunction. Up to 12 months after MI, TK(-/-) mice displayed an increased mortality rate (P<0.05, relative risk of death=3.41) and aggravation of left ventricular hypertrophy and dilatation by comparison with TK(+/+) (+18% and +27% respectively, both P<0.05). NOS1 and NOS3 were abnormally regulated in the heart of TK(-/-) mice after MI. CONCLUSIONS: TK exerts a protective role in HF in mice. Coronary effects are probably involved. As partial genetic deficiency in TK activity occurs in humans, TK-deficient subjects may be at increased risk of mortality in HF.


Asunto(s)
Modelos Animales de Enfermedad , Infarto del Miocardio/fisiopatología , Calicreínas de Tejido/fisiología , Remodelación Ventricular/fisiología , Animales , Circulación Coronaria/fisiología , Ecocardiografía , Cininas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Contracción Miocárdica/fisiología , Infarto del Miocardio/patología , Miocardio/patología , Óxido Nítrico Sintasa/metabolismo , Tamaño de los Órganos , Volumen Sistólico/fisiología , Tasa de Supervivencia , Calicreínas de Tejido/deficiencia
9.
Nephrol Ther ; 14(3): 142-147, 2018 May.
Artículo en Francés | MEDLINE | ID: mdl-29223661

RESUMEN

BACKGROUND AND OBJECTIVES: In France, diabetes mellitus is now the second cause of end stage renal disease. In a large previous French national study, we observed that dialyzed diabetics have a significant lower risk of death by cancer. This first study was focused on cancer death but did not investigate cancer incidence. In this context, the aim of this second study was to compare the incidence of cancer in diabetic dialyzed patients compared to non-diabetic dialyzed patients in a French region. METHODS: This epidemiologic multicentric study included 588 diabetic and non-diabetic patients starting hemodialysis between 2002 and 2007 in Bretagne. Data were issued from REIN registry and cancer incidence were individually collected from medical records. Diabetics and non-diabetics were matched one by one on age, sex and year of dialysis initiation. RESULTS: During the follow-up, we observed 28 cancers (9.4%) in diabetic patients and 26 cancers (8.9%) in non-diabetics patients. The cumulative incidence to develop a cancer 2 years after the dialysis start was approximately 6% in both diabetics and non-diabetics patients. In univariate Fine and Gray analysis, BMI, hemoglobin, statin use had P-value<0.2. However, in the adjusted model, these variables were not significantly associated with cancer incidence. CONCLUSION: This study lead on a little number of dialyzed patients did not show any significant difference on cancer incidence between diabetic and non-diabetic patients after hemodialysis start.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Fallo Renal Crónico/etiología , Neoplasias/epidemiología , Diálisis Renal/efectos adversos , Adulto , Anciano , Femenino , Estudios de Seguimiento , Francia/epidemiología , Humanos , Incidencia , Fallo Renal Crónico/epidemiología , Fallo Renal Crónico/terapia , Masculino , Persona de Mediana Edad , Neoplasias/etiología , Neoplasias/mortalidad , Sistema de Registros , Análisis de Supervivencia
10.
Circulation ; 111(14): 1738-46, 2005 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-15809371

RESUMEN

BACKGROUND: We have previously linked hereditary progressive cardiac conduction defect (hereditary Lenègre's disease) to a loss-of-function mutation in the gene encoding the main cardiac Na+ channel, SCN5A. In the present study, we investigated heterozygous Scn5a-knockout mice (Scn5a+/- mice) as a model for hereditary Lenègre's disease. METHODS AND RESULTS: In Scn5a+/- mice, surface ECG recordings showed age-related lengthening of the P-wave and PR- and QRS-interval duration, coinciding with previous observations in patients with Lenègre's disease. Old but not young Scn5a+/- mice showed extensive fibrosis of their ventricular myocardium, a feature not seen in wild-type animals. In old Scn5a+/- mice, fibrosis was accompanied by heterogeneous expression of connexin 43 and upregulation of hypertrophic markers, including beta-MHC and skeletal alpha-actin. Global connexin 43 expression as assessed with Western blots was similar to wild-type mice. Decreased connexin 40 expression was seen in the atria. Using pangenomic microarrays and real-time PCR, we identified in Scn5a+/- mice an age-related upregulation of genes encoding Atf3 and Egr1 transcription factors. Echocardiography and hemodynamic investigations demonstrated conserved cardiac function with aging and lack of ventricular hypertrophy. CONCLUSIONS: We conclude that Scn5a+/- mice convincingly recapitulate the Lenègre's disease phenotype, including progressive impairment with aging of atrial and ventricular conduction associated with myocardial rearrangements and fibrosis. Our work provides the first demonstration that a monogenic ion channel defect can progressively lead to myocardial structural anomalies.


Asunto(s)
Bloqueo Cardíaco/genética , Sistema de Conducción Cardíaco/fisiopatología , Canales de Sodio/genética , Factores de Edad , Animales , Cardiomegalia , Conexinas/análisis , Modelos Animales de Enfermedad , Electrocardiografía , Fibrosis/genética , Regulación de la Expresión Génica , Enfermedades Genéticas Congénitas , Bloqueo Cardíaco/etiología , Ventrículos Cardíacos/patología , Heterocigoto , Ratones , Ratones Noqueados , Canal de Sodio Activado por Voltaje NAV1.5 , Factores de Transcripción/análisis
11.
Arterioscler Thromb Vasc Biol ; 23(2): 183-9, 2003 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-12588757

RESUMEN

OBJECTIVE: The objective of this study was to investigate the effects of fluvastatin on atherosclerosis, systemic and regional hemodynamics, and vascular reactivity in apolipoprotein E-deficient (ApoE(-/-)) mice. METHODS AND RESULTS: Hemodynamics (fluospheres) and vasomotor responses of thoracic aorta and carotid artery were evaluated in male wild-type (WT) and untreated (ApoE(-/-) Control) or fluvastatin-treated (50 mg/kg per day for 20 weeks) ApoE(-/-) mice, all fed a Western-type diet. Plasma cholesterol and aortic root atherosclerotic lesions (ALs) were greater in ApoE(-/-) Control mice (19+/-1 mmol/L and 63,0176+/-38,785 micro m(2), respectively) than in WT mice (2+/-1 mmol/L and 1+/-1 micro m(2), respectively, P<0.01). Fluvastatin significantly decreased plasma cholesterol (-53%) but failed to limit ALs. Renal blood flow was significantly reduced in ApoE(-/-) Control versus WT (-25%, P<0.05) mice. This reduction was prevented by fluvastatin. Aortic and carotid endothelium-dependent relaxations to acetylcholine were not altered in ApoE(-/-) Control versus WT mice. In carotid arteries from WT mice, these responses were abolished after nitro-L-arginine (L-NA), whereas those from ApoE(-/-) Control were only partially inhibited after L-NA but fully abolished after L-NA+diclofenac. Thus, in carotid arteries from ApoE(-/-) mice, vasodilating prostanoids compensate the deficit in NO availability. Fluvastatin prevented this carotid NO deficit. CONCLUSIONS: In ApoE(-/-) mice, chronic fluvastatin treatment preserved renal perfusion and vascular NO availability independently from atherosclerotic lesion prevention.


Asunto(s)
Apolipoproteínas E/deficiencia , Ácidos Grasos Monoinsaturados/farmacología , Indoles/farmacología , Óxido Nítrico/sangre , Insuficiencia Renal/sangre , Insuficiencia Renal/prevención & control , Acetilcolina/farmacología , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/patología , Aorta Torácica/fisiología , Apolipoproteínas E/genética , Arteriosclerosis/sangre , Arteriosclerosis/metabolismo , Arteriosclerosis/patología , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/patología , Arterias Carótidas/fisiología , Fluvastatina , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Técnicas In Vitro , Riñón/irrigación sanguínea , Riñón/efectos de los fármacos , Lípidos/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Nitroprusiato/farmacología , Perfusión , Resistencia Vascular/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Vasoconstricción/fisiología , Vasoconstrictores/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología , Vasodilatadores/farmacología
12.
J Hypertens ; 20(8): 1581-7, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12172320

RESUMEN

OBJECTIVES: Mechanisms involved in hypertension in homozygous mice for the defective endothelial nitric oxide synthase gene (eNOS-/-) have not been fully elucidated. As NO is a potent vasodilator agent and possibly promotes angiogenesis, we investigated whether vasoconstriction and/or microvascular rarefaction could explain hypertension in these mice. METHODS: Immunohistochemistry with mouse monoclonal smooth muscle alpha-actin antibody was used to detect arterioles, and quantification of arteriolar density was performed in the left ventricle and in the gracilis muscle of 12-week-old male eNOS+/+ and eNOS-/- mice. Haemodynamic parameters - mean arterial pressure (MAP), cardiac index (CI), total peripheral résistance (TPR), myocardial blood flow, muscular blood flow and corresponding resistances - were measured or calculated using the fluorescent microsphere method in basal conditions and after infusion of sodium nitroprusside (SNP) (5 to 150 microg/kg per min) in eNOS-/- mice, compared with eNOS+/+ mice. RESULTS: We evidenced a significant decrease in arteriolar density in the heart (-16%, P < 0.02) and in the gracilis muscle (-22%, P < 0.05) in eNOS-/- mice. In basal conditions, eNOS-/- mice developed significant hypertension (MAP = 127 +/- 14 versus 77 +/- 14 mmHg, P < 0.001) associated with decreased CI (-29%, P < 0.001) and increased TPR (+ 125%, P < 0.001). Coronary and gracilis muscular resistances were increased (by 75 and 89% respectively, P < 0.001) compared with eNOS+/+ mice, whereas myocardial and skeletal muscle tissue blood flows were not affected. After SNP administration (10 microg/kg per min), a dose that did not significantly modify haemodynamic parameters in eNOS+/+ mice, MAP, TPR and regional resistances were normalized in eNOS-/- mice, showing that vasodilation may correct hypertension in eNOS-/- mice. However, under maximal vasodilating conditions, TPR and regional resistances remained significantly higher in eNOS-/- mice than those of eNOS+/+ mice. CONCLUSION: Anatomical and functional results show that both vasoconstriction and arteriolar rarefaction are involved in hypertension of eNOS-/- mice. Indeed, under maximal vasodilation, arterial pressure and TPR remained significantly higher in eNOS-/- mice than in eNOS+/+ mice, evidencing a major role of microvascular rarefaction in this model of hypertension.


Asunto(s)
Hipertensión/etiología , Óxido Nítrico Sintasa/deficiencia , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/genética , Presión Sanguínea/fisiología , Hemodinámica/efectos de los fármacos , Hemodinámica/genética , Hemodinámica/fisiología , Hipertensión/genética , Hipertensión/patología , Hipertensión/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microcirculación/efectos de los fármacos , Microcirculación/patología , Microcirculación/fisiología , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III , Nitroprusiato/farmacología , Resistencia Vascular/efectos de los fármacos , Resistencia Vascular/genética , Resistencia Vascular/fisiología , Vasoconstricción/efectos de los fármacos , Vasoconstricción/genética , Vasoconstricción/fisiología , Vasodilatadores/farmacología
13.
Curr Pharm Des ; 17(25): 2654-62, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21728987

RESUMEN

A fair amount of data indicates that bradykinin and lysyl-bradykinin exert arterial, cardiac and renal effects which afford protection against organ damage in diseases, especially in the settings of ischemia or diabetes. The concept of kinins acting as therapeutic agents is supported by the wide use of angiotensin I-converting enzyme (ACE) inhibitors. These inhibitors indeed potentiate kinin action by inhibiting kinin degradation. Experimental evidence strongly suggests that the cardiac and renal effects of ACE inhibitors are due, at least in part, to kinins. Angiotensin AT1 receptor antagonists act also partly through kinins. This paper reviews available evidence supporting a role for kinins in the therapeutic effect of current drugs. It then discusses the opportunity to develop new drugs based on kinin action. Direct activation of the kinin B2 receptor by pharmacological agonists might provide higher therapeutic benefit than existing kinin- potentiating drugs. Possible occurrence of side effects is however a concern.


Asunto(s)
Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Renales/tratamiento farmacológico , Cininas/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/efectos adversos , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/efectos adversos , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Enfermedades Cardiovasculares/fisiopatología , Sistemas de Liberación de Medicamentos , Diseño de Fármacos , Humanos , Enfermedades Renales/fisiopatología
14.
Am J Physiol Regul Integr Comp Physiol ; 296(5): R1385-91, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19244588

RESUMEN

The kallikrein kinin system (KKS) is involved in arterial and renal functions. It may have an antihypertensive effect in both essential and secondary forms of hypertension. The role of the KKS in the development of two-kidneys, one-clip (2K1C) hypertension, a high-renin model, was investigated in mice rendered deficient in tissue kallikrein (TK) and kinins by TK gene inactivation (TK-/-) and in their wild-type littermates (TK+/+). Four weeks after clipping the renal artery, blood flow was reduced in the clipped kidney (2K1C-TK+/+: -90%, 2K1C-TK-/-: -93% vs. sham-operated mice), and the kidney mass had also decreased (2K1C-TK+/+: -65%, 2K1C-TK-/-: -66%), whereas in the unclipped kidney, blood flow (2K1C-TK+/+: +19%, 2K1C-TK-/-: +17%) and kidney mass (2K1C-TK+/+: +32%, 2K1C-TK-/-: +30%) had both increased. The plasma renin concentration (2K1C-TK+/+: +78%, 2K1C-TK-/-: +65%) and renal renin content of the clipped kidney (2K1C-TK+/+: +58%, 2K1C-TK-/-: +65%) had increased significantly. There was no difference for these parameters between 2K1C-TK+/+ and 2K1C-TK-/- mice. Blood pressure monitored by telemetry and by plethysmography, rose immediately after clipping in both genotypes, and reached similar levels (2K1C-TK+/+: +24%, 2K1C-TK-/-: +21%). 2K1C-TK+/+ and 2K1C-TK-/- mice developed similar concentric left ventricular hypertrophy (+24% and +17%, respectively) with normal cardiac function. These findings suggest that in the context of chronic unilateral reduction in renal blood flow, TK and kinins do not influence the trophicity of kidneys, the synthesis and secretion of renin, blood pressure increase, and cardiac remodeling due to renin angiotensin system activation.


Asunto(s)
Hipertensión Renovascular/metabolismo , Hipertensión Renovascular/fisiopatología , Calicreínas de Tejido/genética , Animales , Presión Sanguínea/fisiología , Vasos Coronarios/fisiología , Modelos Animales de Enfermedad , Hipertensión Renovascular/patología , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/fisiopatología , Riñón/irrigación sanguínea , Riñón/metabolismo , Cininas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Flujo Sanguíneo Regional/fisiología , Arteria Renal/fisiopatología , Renina/metabolismo , Remodelación Ventricular/fisiología
15.
Biol Chem ; 389(6): 701-6, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18627303

RESUMEN

Tissue kallikrein (KLK1) is a kinin-forming serine protease synthesized in many organs including arteries and kidney. Study of the physiological role of KLK1 has benefited from the availability of mouse and human genetic models of KLK1 deficiency, through engineering of KLK1 mouse mutants and discovery of a major polymorphism in the human KLK1 gene that induces a loss of enzyme activity. Studies in KLK1-deficient mice and human subjects partially deficient in KLK1 have documented its critical role in arterial function in both species. KLK1 is also involved in the control of ionic transport in the renal tubule, an action that may not be kinin-mediated. Studies of experimental diseases in KLK1-deficient mice have revealed cardio- and nephro-protective effects of KLK1 and kinins in acute cardiac ischemia, post-ischemic heart failure, and diabetes. Potential clinical and therapeutic developments are discussed.


Asunto(s)
Arterias/fisiología , Corazón/fisiología , Riñón/fisiología , Miocardio/metabolismo , Calicreínas de Tejido/deficiencia , Calicreínas de Tejido/genética , Animales , Arterias/enzimología , Humanos , Riñón/enzimología , Miocardio/enzimología , Miocardio/patología , Calicreínas de Tejido/metabolismo
16.
J Pharmacol Exp Ther ; 323(1): 210-6, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17636004

RESUMEN

Angiotensin-converting enzyme inhibitors limit infarct size in animal models of myocardial ischemia reperfusion injury. This effect has been shown to be due to inhibition of bradykinin degradation rather than inhibition of angiotensin II formation. The purpose of this study was to determine whether angiotensin AT1 receptor blockade by losartan or its active metabolite EXP3174 protects against myocardial ischemia-reperfusion injury in mice and whether this protection is mediated by the kallikrein kinin system. We subjected anesthetized mice to 30 min of coronary artery occlusion followed by 3 h of reperfusion and evaluated infarct size immediately after reperfusion. Losartan (Los) or EXP3174 [2-n-butyl-4-chloro-1-[(2'-(1H-tetrazol-5-yl)biphenyl-4-yI)methyl]imidazole-5-carboxylic acid] were administered 5 min before starting reperfusion at dosages determined by preliminary studies of blood pressure effect and inhibition of angiotensin pressor response. Compared with saline, both drugs significantly reduced myocardial infarct size by roughly 40% (P < 0.001). Pretreatment of mice with the selective AT2 receptor antagonist PD123,319 [S-(+)-1-([4-(dimethylamino)-3-methylphenyl]methyl)-5-(diphenylacetyl)-4,5,6,7-tetrahydro-1H-imidazo(4,5-c)pyridine-6-carboxylic acid] did not affect infarct size in the absence of losartan but abolished the reduction in infarct size provided by losartan. In tissue kallikrein gene-deficient mice (TK-/-), losartan no longer reduced infarct size. Pretreatment of wild-type mice with the B2 receptor antagonist icatibant reproduced the effect of TK deficiency. We conclude that AT1 receptor blockade provides cardioprotection against myocardial ischemia-reperfusion injury through stimulation of AT2 receptors. Kallikrein and B2 receptor are major determinants of this cardioprotective effect of losartan. Our results support the hypothesis of a coupling between AT2 receptors and kallikrein during AT1 receptor blockade, which plays a major role in cardioprotection.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Cardiotónicos/uso terapéutico , Losartán/uso terapéutico , Isquemia Miocárdica/prevención & control , Receptor de Angiotensina Tipo 1/metabolismo , Calicreínas de Tejido/metabolismo , Enfermedad Aguda , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Cardiotónicos/farmacología , Modelos Animales de Enfermedad , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Losartán/farmacología , Ratones , Ratones Noqueados , Isquemia Miocárdica/metabolismo , Calicreínas de Tejido/genética , Función Ventricular Izquierda/efectos de los fármacos
17.
J Cardiovasc Pharmacol ; 42(1): 17-23, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12827021

RESUMEN

We investigated in mice whether atherosclerosis exacerbates the development of post-ischemic heart failure and alters the beneficial effects of long-term angiotensin II type 1 receptor blockade in this model. ApoE-deficient (ApoE(-/-)) and C57BL/6J (C57) mice with myocardial infarction (coronary ligation) received vehicle (C57 and ApoE(-/-)) or irbesartan (Ir, 50mg/kg/d orally, C57-Ir and ApoE(-/-)-Ir). Ten months post myocardial infarction, survival rates were similar in C57 (58%) and ApoE(62%). Atherosclerosis induced no significant alteration in blood pressure, cardiac output (fluospheres), total peripheral resistance, or shortening fraction (echocardiography) but increased renal resistance (+50%, P<0.05). Chronic Ir treatment significantly improved survival to a similar extent in both C57-Ir (85%) and ApoE(-/-)-Ir (86%). It also decreased blood pressure to a similar extent in both strains (-16% and -18%, both P<0.05). In C57-Ir mice, Ir did not modify cardiac output or total peripheral resistance, but it decreased renal resistance (-28%, P<0.001) and left-ventricular weight (-28%, P<0.05). In ApoE(-/-)-Ir mice, Ir limited atherosclerotic lesions (-13%, P<0.05), increased cardiac output (+28%, P<0.05) and shortening fraction (+24%, P<0.05), and decreased total peripheral resistance (-33%, P<0.01), renal resistance (-61%, P<0.001), and left-ventricular weight (-27%, P<0.001). In conclusion, atherosclerosis does not worsen heart failure development in mice and, although the beneficial cardiovascular effects of AT1 receptor blockade are greater in ApoE(-/-) than in C57, reduction in mortality is similar in both strains.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II , Apolipoproteínas E/deficiencia , Compuestos de Bifenilo/uso terapéutico , Infarto del Miocardio/tratamiento farmacológico , Isquemia Miocárdica/metabolismo , Tetrazoles/uso terapéutico , Animales , Arteriosclerosis/complicaciones , Arteriosclerosis/metabolismo , Compuestos de Bifenilo/farmacología , Presión Sanguínea/efectos de los fármacos , Gasto Cardíaco/efectos de los fármacos , Modelos Animales de Enfermedad , Ecocardiografía , Irbesartán , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/etiología , Infarto del Miocardio/metabolismo , Isquemia Miocárdica/complicaciones , Tetrazoles/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA