Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Physiol ; 11: 566871, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33240096

RESUMEN

The kidney is extensively innervated by sympathetic nerves playing an important role in the regulation of blood pressure homeostasis. Sympathetic nerve activity is ultimately controlled by the central nervous system (CNS). Norepinephrine, the main sympathetic neurotransmitter, is released at prejunctional neuroeffector junctions in the kidney and modulates renin release, renal vascular resistance, sodium and water handling, and immune cell response. Under physiological conditions, renal sympathetic nerve activity (RSNA) is modulated by peripheral mechanisms such as the renorenal reflex, a complex interaction between efferent sympathetic nerves, central mechanism, and afferent sensory nerves. RSNA is increased in hypertension and, therefore, critical for the perpetuation of hypertension and the development of hypertensive kidney disease. Renal sympathetic neurotransmission is not only regulated by RSNA but also by prejunctional α2-adrenoceptors. Prejunctional α2-adrenoceptors serve as autoreceptors which, when activated by norepinephrine, inhibit the subsequent release of norepinephrine induced by a sympathetic nerve impulse. Deletion of α2-adrenoceptors aggravates hypertension ultimately by modulating renal pressor response and sodium handling. α2-adrenoceptors are also expressed in the vasculature, renal tubules, and immune cells and exert thereby effects related to vascular tone, sodium excretion, and inflammation. In the present review, we highlight the role of α2-adrenoceptors on renal sympathetic neurotransmission and its impact on hypertension. Moreover, we focus on physiological and pathophysiological functions mediated by non-adrenergic α2-adrenoceptors. In detail, we discuss the effects of sympathetic norepinephrine release and α2-adrenoceptor activation on renal sodium transporters, on renal vascular tone, and on immune cells in the context of hypertension and kidney disease.

2.
J Am Soc Nephrol ; 31(4): 783-798, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32086277

RESUMEN

BACKGROUND: Increased nerve activity causes hypertension and kidney disease. Recent studies suggest that renal denervation reduces BP in patients with hypertension. Renal NE release is regulated by prejunctional α2A-adrenoceptors on sympathetic nerves, and α2A-adrenoceptors act as autoreceptors by binding endogenous NE to inhibit its own release. However, the role of α2A-adrenoceptors in the pathogenesis of hypertensive kidney disease is unknown. METHODS: We investigated effects of α2A-adrenoceptor-regulated renal NE release on the development of angiotensin II-dependent hypertension and kidney disease. In uninephrectomized wild-type and α2A-adrenoceptor-knockout mice, we induced hypertensive kidney disease by infusing AngII for 28 days. RESULTS: Urinary NE excretion and BP did not differ between normotensive α2A-adrenoceptor-knockout mice and wild-type mice at baseline. However, NE excretion increased during AngII treatment, with the knockout mice displaying NE levels that were significantly higher than those of wild-type mice. Accordingly, the α2A-adrenoceptor-knockout mice exhibited a systolic BP increase, which was about 40 mm Hg higher than that found in wild-type mice, and more extensive kidney damage. In isolated kidneys, AngII-enhanced renal nerve stimulation induced NE release and pressor responses to a greater extent in kidneys from α2A-adrenoceptor-knockout mice. Activation of specific sodium transporters accompanied the exaggerated hypertensive BP response in α2A-adrenoceptor-deficient kidneys. These effects depend on renal nerves, as demonstrated by reduced severity of AngII-mediated hypertension and improved kidney function observed in α2A-adrenoceptor-knockout mice after renal denervation. CONCLUSIONS: Our findings reveal a protective role of prejunctional inhibitory α2A-adrenoceptors in pathophysiologic conditions with an activated renin-angiotensin system, such as hypertensive kidney disease, and support the concept of sympatholytic therapy as a treatment.


Asunto(s)
Hipertensión Renal/etiología , Hipertensión Renal/prevención & control , Nefritis/etiología , Nefritis/prevención & control , Receptores Adrenérgicos alfa 2/fisiología , Sistema Nervioso Simpático/fisiopatología , Transmisión Sináptica/fisiología , Angiotensina II , Animales , Modelos Animales de Enfermedad , Hipertensión Renal/fisiopatología , Ratones , Ratones Noqueados , Nefritis/fisiopatología , Simpatectomía
3.
Circulation ; 139(11): 1407-1421, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30586752

RESUMEN

BACKGROUND: Arterial hypertension and its organ sequelae show characteristics of T cell-mediated inflammatory diseases. Experimental anti-inflammatory therapies have been shown to ameliorate hypertensive end-organ damage. Recently, the CANTOS study (Canakinumab Antiinflammatory Thrombosis Outcome Study) targeting interleukin-1ß demonstrated that anti-inflammatory therapy reduces cardiovascular risk. The gut microbiome plays a pivotal role in immune homeostasis and cardiovascular health. Short-chain fatty acids (SCFAs) are produced from dietary fiber by gut bacteria and affect host immune homeostasis. Here, we investigated effects of the SCFA propionate in 2 different mouse models of hypertensive cardiovascular damage. METHODS: To investigate the effect of SCFAs on hypertensive cardiac damage and atherosclerosis, wild-type NMRI or apolipoprotein E knockout-deficient mice received propionate (200 mmol/L) or control in the drinking water. To induce hypertension, wild-type NMRI mice were infused with angiotensin II (1.44 mg·kg-1·d-1 subcutaneous) for 14 days. To accelerate the development of atherosclerosis, apolipoprotein E knockout mice were infused with angiotensin II (0.72 mg·kg-1·d-1 subcutaneous) for 28 days. Cardiac damage and atherosclerosis were assessed using histology, echocardiography, in vivo electrophysiology, immunofluorescence, and flow cytometry. Blood pressure was measured by radiotelemetry. Regulatory T cell depletion using PC61 antibody was used to examine the mode of action of propionate. RESULTS: Propionate significantly attenuated cardiac hypertrophy, fibrosis, vascular dysfunction, and hypertension in both models. Susceptibility to cardiac ventricular arrhythmias was significantly reduced in propionate-treated angiotensin II-infused wild-type NMRI mice. Aortic atherosclerotic lesion area was significantly decreased in propionate-treated apolipoprotein E knockout-deficient mice. Systemic inflammation was mitigated by propionate treatment, quantified as a reduction in splenic effector memory T cell frequencies and splenic T helper 17 cells in both models, and a decrease in local cardiac immune cell infiltration in wild-type NMRI mice. Cardioprotective effects of propionate were abrogated in regulatory T cell-depleted angiotensin II-infused mice, suggesting the effect is regulatory T cell-dependent. CONCLUSIONS: Our data emphasize an immune-modulatory role of SCFAs and their importance for cardiovascular health. The data suggest that lifestyle modifications leading to augmented SCFA production could be a beneficial nonpharmacological preventive strategy for patients with hypertensive cardiovascular disease.


Asunto(s)
Antiinflamatorios/farmacología , Enfermedades de la Aorta/tratamiento farmacológico , Arritmias Cardíacas/prevención & control , Aterosclerosis/tratamiento farmacológico , Cardiomegalia/prevención & control , Hipertensión/tratamiento farmacológico , Propionatos/farmacología , Angiotensina II , Animales , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/patología , Arritmias Cardíacas/inmunología , Arritmias Cardíacas/fisiopatología , Presión Arterial/efectos de los fármacos , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/patología , Cardiomegalia/inmunología , Cardiomegalia/fisiopatología , Modelos Animales de Enfermedad , Hipertensión/inducido químicamente , Hipertensión/inmunología , Hipertensión/fisiopatología , Masculino , Ratones Noqueados para ApoE , Placa Aterosclerótica , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Células Th17/efectos de los fármacos , Células Th17/inmunología
4.
Int J Mol Sci ; 19(4)2018 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-29570672

RESUMEN

Nitric oxide (NO) modulates renal blood flow (RBF) and kidney function and is involved in blood pressure (BP) regulation predominantly via stimulation of the NO-sensitive guanylyl cyclase (NO-GC), existing in two isoforms, NO-GC1 and NO-GC2. Here, we used isoform-specific knockout (KO) mice and investigated their contribution to renal hemodynamics under normotensive and angiotensin II-induced hypertensive conditions. Stimulation of the NO-GCs by S-nitrosoglutathione (GSNO) reduced BP in normotensive and hypertensive wildtype (WT) and NO-GC2-KO mice more efficiently than in NO-GC1-KO. NO-induced increase of RBF in normotensive mice did not differ between the genotypes, but the respective increase under hypertensive conditions was impaired in NO-GC1-KO. Similarly, inhibition of endogenous NO increased BP and reduced RBF to a lesser extent in NO-GC1-KO than in NO-GC2-KO. These findings indicate NO-GC1 as a target of NO to normalize RBF in hypertension. As these effects were not completely abolished in NO-GC1-KO and renal cyclic guanosine monophosphate (cGMP) levels were decreased in both NO-GC1-KO and NO-GC2-KO, the results suggest an additional contribution of NO-GC2. Hence, NO-GC1 plays a predominant role in the regulation of BP and RBF, especially in hypertension. However, renal NO-GC2 appears to compensate the loss of NO-GC1, and is able to regulate renal hemodynamics under physiological conditions.


Asunto(s)
Guanilil Ciclasa Soluble/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , GMP Cíclico/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Ratones , Ratones Noqueados , NG-Nitroarginina Metil Éster/metabolismo , Óxido Nítrico/metabolismo , Circulación Renal/efectos de los fármacos , S-Nitrosoglutatión/farmacología , Vasodilatación/efectos de los fármacos
5.
Pflugers Arch ; 470(4): 661-667, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29352340

RESUMEN

Angiotensin (Ang)-(1-7) ameliorates vascular injury by increasing nitric oxide (NO) bioavailability. Evidence that Ang-(1-7) attenuates the development of atherosclerosis through a NO-dependent mechanism is still missing. Moreover, it has been postulated that Ang-(1-7) may mediate its effects by other mechanisms than Mas receptor activation. To investigate Ang-(1-7)-dependent Mas receptor function, we treated apoE-KO and apoE/Mas-KO mice chronically with Ang-(1-7) (82 µg/kg per hour) or saline for 6 weeks. Flow-mediated dilation (FMD), a measure for NO-dependent vasodilation and the most accepted prognostic marker for the development of atherosclerosis, was measured in vivo. Chronic Ang-(1-7) treatment improved FMD and attenuated the development of atherosclerosis in apolipoproteinE (apoE)-KO but not in apoE/Mas-KO mice. These effects were accompanied by increased aortic nitrite and cGMP levels. To test whether Ang-(1-7) modulates atherosclerosis through a NO-dependent mechanism, apoE-KO mice were treated with the NO synthase inhibitor L-NAME (20 mg/kg/day) in the presence or absence of Ang-(1-7). L-NAME treatment reduced aortic nitrite content and increased blood pressure and exaggerated atherosclerosis compared to untreated apoE-KO mice. In L-NAME-treated apoE-KO mice, chronic Ang-(1-7) treatment did not increase aortic nitrite content and consequently showed no effect on blood pressure and the development of atherosclerosis. The present study proves that Ang-(1-7) mediates its protective vascular effects through Mas receptor activation. Moreover, Ang-(1-7)-mediated NO generation is essential for improving vascular function and prevents atherosclerosis in apoE-KO mice.


Asunto(s)
Angiotensina I/farmacología , Apolipoproteínas E/genética , Aterosclerosis/tratamiento farmacológico , Óxido Nítrico/metabolismo , Fragmentos de Péptidos/farmacología , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Aterosclerosis/metabolismo , Presión Sanguínea/efectos de los fármacos , GMP Cíclico/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/metabolismo , Proto-Oncogenes Mas , Vasodilatación/efectos de los fármacos
6.
Am J Physiol Renal Physiol ; 312(3): F474-F481, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28052870

RESUMEN

Changes in renal hemodynamics have a major impact on blood pressure (BP). Angiotensin (Ang) II has been shown to induce vascular dysfunction by interacting with phosphodiesterase (PDE)1 and PDE5. The predominant PDE isoform responsible for renal vascular dysfunction in hypertension is unknown. Here, we measured the effects of PDE5 (sildenafil) or PDE1 (vinpocetine) inhibition on renal blood flow (RBF), BP, and renal vascular function in normotensive and hypertensive mice. During acute short-term Ang II infusion, sildenafil decreased BP and increased RBF in C57BL/6 (WT) mice. In contrast, vinpocetine showed no effect on RBF and BP. Additionally, renal cGMP levels were significantly increased after acute sildenafil but not after vinpocetine infusion, indicating a predominant role of PDE5 in renal vasculature. Furthermore, chronic Ang II infusion (500 ng·kg-1·min-1) increased BP and led to impaired NO-dependent vasodilation in kidneys of WT mice. Additional treatment with sildenafil (100 mg·kg-1·day-1) attenuated Ang II-dependent hypertension and improved NO-mediated vasodilation. During chronic Ang II infusion, urinary nitrite excretion, a marker for renal NO generation, was increased in WT mice, whereas renal cGMP generation was decreased and restored after sildenafil treatment, suggesting a preserved cGMP signaling after PDE5 inhibition. To investigate the dependency of PDE5 effects on NO/cGMP signaling, we next analyzed eNOS-KO mice, a mouse model characterized by low vascular NO/cGMP levels. In eNOS-KO mice, chronic Ang II infusion increased BP but did not impair NO-mediated vasodilation. Moreover, sildenafil did not influence BP or vascular function in eNOS-KO mice. These results highlight PDE5 as a key regulator of renal hemodynamics in hypertension.


Asunto(s)
Angiotensina II , Antihipertensivos/farmacología , Presión Sanguínea/efectos de los fármacos , Hipertensión/prevención & control , Inhibidores de Fosfodiesterasa 5/farmacología , Arteria Renal/efectos de los fármacos , Circulación Renal/efectos de los fármacos , Citrato de Sildenafil/farmacología , Vasodilatadores/farmacología , Animales , GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Hipertensión/inducido químicamente , Hipertensión/enzimología , Hipertensión/fisiopatología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/deficiencia , Óxido Nítrico Sintasa de Tipo III/genética , Arteria Renal/enzimología , Arteria Renal/fisiopatología , Vasodilatación/efectos de los fármacos , Alcaloides de la Vinca/farmacología
7.
Hypertension ; 59(2): 395-401, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22184318

RESUMEN

Preeclampsia is associated with increased risk of cardiovascular disease. Midregional proatrial natriuretic peptide (MR-proANP), a precursor of the atrial natriuretic peptide, is a biomarker for cardiovascular disease. We obtained plasma from 184 pregnant women in gestational weeks 24 to 42 (normotensive pregnancies: n=77, preeclampsia: n=107), from 25 of these women at 5 to 8 years after index pregnancy (normotensive pregnancies: n=11, preeclampsia: n=14), and from 49 normotensive, nonpregnant women and analyzed them by immunoassay for MR-proANP. To investigate potential sources, placental and decidual atrial natriuretic peptide mRNA expression levels were analyzed by quantitative real-time PCR in 21 normotensive and 23 preeclamptic pregnancies, as well as in human heart and kidney samples. For further confirmation, we measured circulating MR-proANP and performed expression studies in a transgenic rat model for preeclampsia. MR-proANP was significantly elevated in maternal plasma in preeclampsia compared with normotensive pregnancies (135 versus 56 pmol/L; P<0.001). However, 5 to 8 years after pregnancy, there was no difference (formerly preeclamptic women versus formerly normotensive in pregnancy: 53 versus 49 pmol/L; P=0.5). Our preeclamptic rat model confirmed the acute MR-proANP differences between preeclamptic and normotensive pregnancies (10.9±1.9 versus 4.3±0.3 pmol/L; P=0.05). Atrial natriuretic peptide expression was high in the heart but negligible in the uteroplacental unit in both normotensive humans and rats, whereas expression in maternal and fetal hearts in the preeclamptic rats was significantly increased, compared with controls. MR-proANP is a serviceable biomarker in preeclampsia, both in humans and a rat model, probably reflecting cardiovascular hemodynamic stress.


Asunto(s)
Factor Natriurético Atrial/metabolismo , Preeclampsia/metabolismo , Segundo Trimestre del Embarazo/metabolismo , Tercer Trimestre del Embarazo/metabolismo , Adulto , Animales , Biomarcadores/metabolismo , Enfermedades Cardiovasculares/epidemiología , Modelos Animales de Enfermedad , Femenino , Humanos , Riñón/metabolismo , Miocardio/metabolismo , Embarazo , ARN Mensajero/metabolismo , Ratas , Ratas Transgénicas , Factores de Riesgo
8.
Hypertension ; 58(1): 77-84, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21576625

RESUMEN

Pregnant women who subsequently develop preeclampsia are highly sensitive to infused angiotensin (Ang) II; the sensitivity persists postpartum. Activating autoantibodies against the Ang II type 1 (AT(1)) receptor are present in preeclampsia. In vitro and in vivo data suggest that they could be involved in the disease process. We generated and purified activating antibodies against the AT(1) receptor (AT(1)-AB) by immunizing rabbits against the AFHYESQ epitope of the second extracellular loop, which is the binding epitope of endogenous activating autoantibodies against AT(1) from patients with preeclampsia. We then purified AT(1)-AB using affinity chromatography with the AFHYESQ peptide. We were able to detect AT(1)-AB both by ELISA and a functional bioassay. We then passively transferred AT(1)-AB into pregnant rats, alone or combined with Ang II. AT(1)-AB activated protein kinase C-α and extracellular-related kinase 1/2. Passive transfer of AT(1)-AB alone or Ang II (435 ng/kg per minute) infused alone did not induce a preeclampsia-like syndrome in pregnant rats. However, the combination (AT(1)-AB plus Ang II) induced hypertension, proteinuria, intrauterine growth retardation, and arteriolosclerosis in the uteroplacental unit. We next performed gene-array profiling of the uteroplacental unit and found that hypoxia-inducible factor 1α was upregulated by Ang II plus AT(1)-AB, which we then confirmed by Western blotting in villous explants. Furthermore, endothelin 1 was upregulated in endothelial cells by Ang II plus AT(1)-AB. We show that AT(1)-AB induces Ang II sensitivity. Our mechanistic study supports the existence of an "autoimmune-activating receptor" that could contribute to Ang II sensitivity and possible to preeclampsia.


Asunto(s)
Angiotensina II/genética , Autoanticuerpos , Regulación del Desarrollo de la Expresión Génica , Preeclampsia/inmunología , Preñez , ARN/genética , Receptor de Angiotensina Tipo 1/inmunología , Angiotensina II/metabolismo , Animales , Western Blotting , Células Cultivadas , Cricetinae , Ensayo de Inmunoadsorción Enzimática , Femenino , Feto/embriología , Feto/metabolismo , Preeclampsia/genética , Preeclampsia/metabolismo , Embarazo , Conejos , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1/metabolismo
9.
Hypertension ; 56(2): 304-10, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20606107

RESUMEN

Rats harboring the human angiotensinogen and human renin genes develop preeclamptic features in pregnancy. The preeclamptic rats exhibit a deeper trophoblast invasion associated with a reduced resistance index by uterine Doppler. Doxycycline inhibits matrix metalloproteinase activity. We tested the hypothesis that matrix metalloproteinase inhibition reduces trophoblast invasion with subsequent changes in placental perfusion. Preeclamptic and pregnant control Sprague-Dawley rats were treated with doxycycline (30 mg/kg of body weight orally) from gestational day 12 until day 18. Placental perfusion was assessed using a micromarker contrast agent. The animals were euthanized on day 18 of pregnancy; biometric data were acquired, and trophoblast invasion was analyzed. Doxycycline resulted in intrauterine growth retardation and lighter placentas in both groups. Maternal body weight was not affected. As shown earlier, preeclamptic rats exhibited a deeper endovascular trophoblast invasion. However, doxycycline treatment reduced trophoblast invasion in the preeclamptic rats. The physiological spiral artery remodeling, as assessed by the deposition of fibrinoid and alpha-actin in the spiral artery contour, was significantly reduced by doxycycline. The vascularity index, as assessed by perfusion measurement of the placenta, was reduced after doxycycline treatment in preeclamptic rats. Thus, matrix metalloproteinase inhibition with doxycycline leads to reduced trophoblast invasion and associated reduced placental perfusion. These studies are the first to show that reducing trophoblast-induced vascular remodeling decreases subsequent placental perfusion. Our model allows the study of dysregulated trophoblast invasion and vascular remodeling in vivo to gain important insights into preeclampsia-related mechanisms.


Asunto(s)
Arterias/fisiología , Placenta/irrigación sanguínea , Placenta/fisiología , Trofoblastos/fisiología , Angiotensinógeno/genética , Animales , Apoptosis/fisiología , Presión Sanguínea , Femenino , Humanos , Tamaño de los Órganos , Preeclampsia/genética , Preeclampsia/fisiopatología , Embarazo , Ratas , Ratas Sprague-Dawley , Valores de Referencia , Renina/genética , Sístole/fisiología , Trofoblastos/patología
10.
Hypertension ; 56(2): 311-8, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20530295

RESUMEN

The renin-angiotensin (Ang) system is important during placental development. Dysregulation of the renin-Ang system is important in preeclampsia (PE). Female rats transgenic for the human angiotensinogen gene crossed with males transgenic for the human renin gene develop the PE syndrome, whereas those of the opposite cross do not. We used this model to study the role of Ang II in trophoblast invasion, which is shallow in human PE but deeper in this model. We investigated the following groups: PE rats, opposite-cross rats, Ang II-infused rats (1000 ng/kg per day), and control rats. Ang II infusion increased only circulating Ang II levels (267.82 pg/mL), opposite cross influenced only uteroplacental Ang II (13.52 fmol/mg of protein), and PE increased both circulating (251.09 pg/mL) and uteroplacental (19.24 fmol/mg of protein) Ang II. Blood pressure and albuminuria occurred in the models with high circulating Ang II but not in the other models. Trophoblast invasion increased in PE and opposite-cross rats but not in Ang II-infused rats. Correspondingly, uterine artery resistance index increased in Ang II-infused rats but decreased in PE rats. We then studied human trophoblasts and villous explants from first-trimester pregnancies with time-lapse microscopy. Local Ang II dose-dependently increased migration by 75%, invasion by 58%, and motility by 282%. The data suggest that local tissue Ang II stimulates trophoblast invasion in vivo in the rat and in vitro in human cells, a hitherto fore unrecognized function. Conceivably, upregulation of tissue Ang II in the maternal part of the placenta represents an important growth factor for trophoblast invasion and migration.


Asunto(s)
Angiotensina II/fisiología , Angiotensinógeno/genética , Retardo del Crecimiento Fetal/genética , Placenta/fisiología , Preeclampsia/genética , Preñez , Renina/genética , Útero/fisiología , Angiotensina II/sangre , Angiotensina II/farmacología , Animales , Animales Modificados Genéticamente , Encéfalo/anatomía & histología , Encéfalo/embriología , Movimiento Celular/fisiología , Cruzamientos Genéticos , Femenino , Humanos , Hígado/anatomía & histología , Hígado/embriología , Masculino , Embarazo , Preñez/fisiología , Ratas , Trofoblastos/efectos de los fármacos , Trofoblastos/patología , Regulación hacia Arriba
11.
Hypertension ; 55(2): 562-6, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20038747

RESUMEN

The pregnant female human angiotensinogen (hAGN) transgenic rat mated with the male hrenin (hREN) transgenic rat is a model of preeclampsia with increased blood pressure, proteinuria, and placenta alterations of edema and necrosis. The reverse mating of female hRENxmale hAGN does not show preeclamptic features. Because the placenta is well-recognized to be a key contributor to the preeclamptic syndrome, our hypothesis is that local angiotensin peptide concentrations found in the placenta and its associated mesometrial triangle of the preeclamptic transgenic rat differ from the reverse mating. We characterized the angiotensin peptide content and the mRNA expression of hREN and hAGN of the mesometrial triangle and the placenta. Three groups of pregnant rats from the matings (Sprague-DawleyxSprague-Dawley, reverse mating, and female hAGNxmale hREN) were studied on day 21 of gestation. In the hAGNxhREN transgenic rat, angiotensin II is significantly increased in the placenta and mesometrial triangle vs Sprague-Dawley (24.2+/-3.9 vs 8.6+/-1.5 pg/mg protein; 27.8+/-5.5 vs 5.6+/-1.3 pg/mg protein; P<0.05), whereas in the reverse mating angiotensin II is increased in the placenta (19.1+/-1.7 vs 5.6+/-1.3 pg/mg protein; P<0.05) but unchanged in the mesometrial triangle (4.2+/-0.2 vs 8.6+/-1.5 pg/mg protein). The marked contrast in the expression of angiotensin II in the mesometrial triangle of the preeclamptic model vs the reverse mating suggests that local angiotensin II generated from the maternal parts of the uteroplacental unit may play a critical role in preeclampsia.


Asunto(s)
Angiotensina II/metabolismo , Placenta/metabolismo , Preeclampsia/metabolismo , Sistema Renina-Angiotensina/fisiología , Análisis de Varianza , Animales , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Humanos , Preeclampsia/genética , Embarazo , Probabilidad , ARN Mensajero/análisis , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas
12.
J Mol Med (Berl) ; 86(6): 697-703, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18398593

RESUMEN

Pre-eclampsia is a common, pregnancy-induced disorder, consisting of hypertension and proteinuria. The condition is one of the leading causes for maternal and perinatal morbidity and mortality. Nonetheless, the underlying molecular mechanisms remain unclear. Immunological mechanisms and the renin-angiotensin system have been implicated in the development of pre-eclampsia. Agonistic autoantibodies to the angiotensin II type I receptor (AT1-AA) have been identified in pre-eclamptic patients, unifying the two hypothesis. Evidence has also accumulated for the existence and importance of a local, utroplacental renin-angiotensin system. We summarize recent data emphasizing the pathophysiological role for the autoantibodies and the uteroplacental renin-angiotensin system.


Asunto(s)
Autoanticuerpos/inmunología , Circulación Placentaria/inmunología , Preeclampsia/inmunología , Receptor de Angiotensina Tipo 1/inmunología , Sistema Renina-Angiotensina/inmunología , Animales , Femenino , Humanos , Preeclampsia/patología , Embarazo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
13.
Hypertension ; 51(2): 547-53, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18195162

RESUMEN

We investigated intrauterine growth restriction, endothelial function, and uterine artery blood flow characteristics in a transgenic preeclampsia rat model with an activated renin-angiotensin system. We compared preeclamptic Sprague-Dawley (SD-PE) rats with normal pregnant Sprague-Dawley and nonpregnant Sprague-Dawley rats. We used transabdominal ultrasound and found that SD-PE rat embryos developed intrauterine growth restriction. Isolated uterine arteries from SD-PE rats incubated with phenylephrine exhibited an increased contractile response, whereas a single high dose of acetylcholine resulted in an impaired vasorelaxation compared with controls. Incremental acetylcholine doses increased relaxation of SD-PE vessels at low acetylcholine doses but caused a paradoxical contraction at higher acetylcholine doses. Indomethacin and a thromboxane-receptor antagonist (SQ 29,548) blocked this effect, suggesting maternal prostanoid-dependent endothelial dysfunction. SD-PE rats had a decreased prostacyclin (6-keto-prostaglandin F1alpha):thromboxane ratio in the serum compared with normal pregnant Sprague-Dawley rats or nonpregnant Sprague-Dawley. Surprisingly, the Doppler resistance index decreased during pregnancy in SD-PE compared with normal pregnant Sprague-Dawley rats, suggesting unimpaired uteroplacental flow in the uterine artery. Umbilical flow was unchanged with absent end-diastolic flow in all of the groups. Renin-angiotensin system activation-induced preeclampsia is associated with altered placentation, modified resistance index, and endothelial dysfunction. A disturbed prostacyclin:thromboxane ratio could be an important mediator.


Asunto(s)
Preeclampsia/fisiopatología , Útero/irrigación sanguínea , Acetilcolina/administración & dosificación , Acetilcolina/farmacología , Animales , Animales Modificados Genéticamente , Arterias/efectos de los fármacos , Arterias/fisiopatología , Vasos Sanguíneos/diagnóstico por imagen , Vasos Sanguíneos/fisiopatología , Relación Dosis-Respuesta a Droga , Endotelio Vascular/fisiopatología , Epoprostenol/sangre , Femenino , Retardo del Crecimiento Fetal/diagnóstico por imagen , Retardo del Crecimiento Fetal/etiología , Técnicas In Vitro , Masculino , Fenilefrina/farmacología , Preeclampsia/genética , Preeclampsia/metabolismo , Embarazo , Ratas , Ratas Sprague-Dawley , Flujo Sanguíneo Regional , Sistema Renina-Angiotensina , Tromboxanos/sangre , Ultrasonografía Prenatal , Resistencia Vascular , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología , Vasodilatadores/farmacología
14.
Hypertension ; 51(2): 554-9, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18195163

RESUMEN

Maternal spiral artery remodeling is the consequence of controlled trophoblast invasive interaction with the maternal cellular environment and is fundamentally important for successful placentation. In preeclampsia, trophoblast invasion is shallow, remodeling is incomplete, and vessels develop an inflammatory appearance, termed "acute atherosis." We noted that, in our preeclampsia, human renin-human angiotensinogen transgenic rat model, complement component 3 (C3), and tumor necrosis factor-alpha were upregulated and heavily expressed in atherotic uteroplacental vessels. We next used coculture involving human trophoblasts, rat vascular smooth muscle cells (VSMCs), and human VSMCs to observe VSMC-trophoblast regulatory interactions. Tumor necrosis factor-alpha induced complement C3 and interleukin-6 expression in VSMCs. We found that trophoblasts were able to reduce VSMC C3 and interleukin-6 expression after the VSMCs were stimulated with tumor necrosis factor-alpha. However, a direct VSMC-trophoblast cell-cell contact was necessary for this anti-inflammatory response. We also studied double-transgenic VSMCs that express inflammatory components and exhibit accelerated proliferation ("synthetic" phenotype). Trophoblasts could not downregulate C3 in these cells. We then examined uteroplacental tissues from preeclamptic and control patients. In control deciduas, only traces of C3 staining were observed, and vessels were thin walled without thrombus formation. In preeclampsia, the decidual vessels showed atherosis, thrombus formation, and C3 expression. Our data suggest that fetally derived trophoblasts require direct cell-cell contact with maternally derived VSMCs to downregulate VSMC C3 and interleukin-6 expression and to avoid atherosis. The findings also implicate C3 in the placental vasculopathy observed in preeclampsia.


Asunto(s)
Aterosclerosis/prevención & control , Comunicación Celular/fisiología , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/fisiología , Preeclampsia/fisiopatología , Trofoblastos/fisiología , Enfermedad Aguda , Angiotensinógeno/genética , Angiotensinógeno/metabolismo , Animales , Animales Modificados Genéticamente , Aterosclerosis/etiología , Células Cultivadas , Vellosidades Coriónicas , Técnicas de Cocultivo , Complemento C3/metabolismo , Decidua/irrigación sanguínea , Femenino , Humanos , Masculino , Preeclampsia/metabolismo , Embarazo , Ratas , Ratas Sprague-Dawley , Renina/genética , Renina/metabolismo , Trombosis/etiología
15.
Hypertension ; 49(3): 604-11, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17261642

RESUMEN

The renin-angiotensin system (RAS) participates in preeclampsia; however, the relative contributions from the circulating RAS and the tissue-based, uteroplacental RAS are unknown. We hypothesized that the tissue-based uteroplacental RAS is dysregulated in preeclampsia. We performed microarray and gene expression studies and confirmed the findings on the protein level by immunohistochemistry in ureteroplacental units from 10 preeclamptic women and 10 women with uneventful pregnancies. All of the women were delivered by cesarean section. We also analyzed plasma renin activity and circulating agonistic angiotensin II type 1 (AT1) receptor autoantibodies. In preeclampsia, we found that the angiotensin II AT1 receptor gene was 5-fold upregulated in decidua (maternal origin). We also found AT1 autoantibodies in preeclamptic women and in their offspring by neonatal cardiomyocyte bioassay compared with women with normal pregnancies and their infants (mother: 17.5+/-2.2 versus 0.05+/-0.4; fetus: 14.5+/-1.8 versus 0.5+/-0.5 Deltabpm). Gene expressions for renin (35.0-fold), angiotensin-converting enzyme (2.9-fold), and angiotensinogen (8.9-fold) were higher in decidua than placenta (fetal origin) in both control and preeclamptic women, whereas the AT1 receptor was expressed 10-fold higher in placenta than in decidua in both groups. Our findings elucidate the ureteroplacental unit RAS in preeclamptic and normal pregnancies. We found that, in preeclampsia, the AT1 receptor expression is particularly high in decidua, combined with pregnancy-specific tissue RAS involving decidual angiotensin II production and AT1 autoantibodies. We also showed that AT1 autoantibodies cross the ureteroplacental barrier. These components could participate in the pathophysiology of preeclampsia.


Asunto(s)
Preeclampsia/fisiopatología , Sistema Renina-Angiotensina/fisiología , Adulto , Angiotensinas/análisis , Angiotensinas/biosíntesis , Autoanticuerpos , Decidua/química , Decidua/fisiopatología , Femenino , Humanos , Placenta/química , Placenta/fisiopatología , Circulación Placentaria , Preeclampsia/genética , Embarazo , Receptor de Angiotensina Tipo 1/biosíntesis , Receptor de Angiotensina Tipo 1/inmunología , Renina/análisis , Renina/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA