Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Mol Cell Endocrinol ; 529: 111255, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33789143

RESUMEN

The renin-angiotensin system (RAS) is a highly complex hormonal cascade that spans multiple organs and cell types to regulate solute and fluid balance along with cardiovascular function. Much of our current understanding of the functions of the RAS has emerged from a series of key studies in genetically-modified animals. Here, we review key findings from ground-breaking transgenic models, spanning decades of research into the RAS, with a focus on their use in studying blood pressure. We review the physiological importance of this regulatory system as evident through the examination of mouse models for several major RAS components: angiotensinogen, renin, ACE, ACE2, and the type 1 A angiotensin receptor. Both whole-animal and cell-specific knockout models have permitted critical RAS functions to be defined and demonstrate how redundancy and multiplicity within the RAS allow for compensatory adjustments to maintain homeostasis. Moreover, these models present exciting opportunities for continued discovery surrounding the role of the RAS in disease pathogenesis and treatment for cardiovascular disease and beyond.


Asunto(s)
Angiotensinógeno/genética , Enfermedades Cardiovasculares/genética , Modelos Animales de Enfermedad , Sistema Renina-Angiotensina/genética , Renina/genética , Equilibrio Hidroelectrolítico/genética , Enzima Convertidora de Angiotensina 2/deficiencia , Enzima Convertidora de Angiotensina 2/genética , Angiotensinógeno/deficiencia , Animales , Presión Sanguínea/genética , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Regulación de la Expresión Génica , Humanos , Riñón/citología , Riñón/metabolismo , Ratones , Ratones Noqueados , Receptor de Angiotensina Tipo 1/deficiencia , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 2/deficiencia , Receptor de Angiotensina Tipo 2/genética , Renina/deficiencia , Transducción de Señal
3.
Arterioscler Thromb Vasc Biol ; 40(9): 2108-2113, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32640904

RESUMEN

OBJECTIVE: Renin cleavage of angiotensinogen has species specificity. As the residues at positions 11 and 12 are different between human angiotensinogen and mouse angiotensinogen, we determined whether these 2 residues in angiotensinogen affect renin cleavage and angiotensin II-mediated blood pressure regulation and atherosclerosis using an adenoassociated viral approach for manipulating angiotensinogen in vivo. Approach and Results: Hepatocyte-specific angiotensinogen deficient (hepAGT-/-) mice in an LDL receptor-deficient background were infected with adenoassociated virals containing a null insert, human angiotensinogen, or mouse angiotensinogen expressing the same residues of the human protein at positions 11 and 12 (mouse angiotensinogen [L11V;Y12I]). Expression of human angiotensinogen in hepAGT-/- mice led to high plasma human angiotensinogen concentrations without changes in plasma endogenous mouse angiotensinogen, plasma renin concentrations, blood pressure, or atherosclerosis. This is consistent with human angiotensinogen not being cleaved by mouse renin. To determine whether the residues at positions 11 and 12 in human angiotensinogen lead to the inability of mouse renin to cleave human angiotensinogen, hepAGT-/- mice were injected with adenoassociated viral vector encoding mouse angiotensinogen (L11V;Y12I). Expression of mouse angiotensinogen (L11V;Y12I) in hepAGT-/- mice resulted in increased plasma mouse angiotensinogen concentrations, reduced renin concentrations, and increased renal AngII concentrations that were comparable to their concentrations in hepAGT+/+ mice. This mouse angiotensinogen variant increased blood pressure and atherosclerosis in hepAGT-/- mice to the magnitude of hepAGT+/+ mice. CONCLUSIONS: Replacement of L11 and Y12 to V11 and I12, respectively, in mouse angiotensinogen does not affect renin cleavage, blood pressure, and atherosclerosis in LDL receptor-deficient mice.


Asunto(s)
Angiotensina II/metabolismo , Angiotensinógeno/metabolismo , Aterosclerosis/metabolismo , Presión Sanguínea , Hepatocitos/metabolismo , Hipertensión/metabolismo , Renina/metabolismo , Sustitución de Aminoácidos , Angiotensinógeno/deficiencia , Angiotensinógeno/genética , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Hipertensión/genética , Hipertensión/fisiopatología , Masculino , Ratones Noqueados , Placa Aterosclerótica , Receptores de LDL/genética , Receptores de LDL/metabolismo , Especificidad de la Especie
4.
Am J Physiol Regul Integr Comp Physiol ; 318(5): R855-R869, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32186897

RESUMEN

Angiotensin II (ANG II) Agtr1a receptor (AT1A) is expressed in cells of the arcuate nucleus of the hypothalamus that express the leptin receptor (Lepr) and agouti-related peptide (Agrp). Agtr1a expression in these cells is required to stimulate resting energy expenditure in response to leptin and high-fat diets (HFDs), but the mechanism activating AT1A signaling by leptin remains unclear. To probe the role of local paracrine/autocrine ANG II generation and signaling in this mechanism, we bred mice harboring a conditional allele for angiotensinogen (Agt, encoding AGT) with mice expressing Cre-recombinase via the Lepr or Agrp promoters to cause cell-specific deletions of Agt (AgtLepr-KO and AgtAgrp-KO mice, respectively). AgtLepr-KO mice were phenotypically normal, arguing against a paracrine/autocrine AGT signaling mechanism for metabolic control. In contrast, AgtAgrp-KO mice exhibited reduced preweaning survival, and surviving adults exhibited altered renal structure and steroid flux, paralleling previous reports of animals with whole body Agt deficiency or Agt disruption in albumin (Alb)-expressing cells (thought to cause liver-specific disruption). Surprisingly, adult AgtAgrp-KO mice exhibited normal circulating AGT protein and hepatic Agt mRNA expression but reduced Agt mRNA expression in adrenal glands. Reanalysis of RNA-sequencing data sets describing transcriptomes of normal adrenal glands suggests that Agrp and Alb are both expressed in this tissue, and fluorescent reporter gene expression confirms Cre activity in adrenal gland of both Agrp-Cre and Alb-Cre mice. These findings lead to the iconoclastic conclusion that extrahepatic (i.e., adrenal) expression of Agt is critically required for normal renal development and survival.


Asunto(s)
Glándulas Suprarrenales/metabolismo , Proteína Relacionada con Agouti/metabolismo , Angiotensinógeno/metabolismo , Núcleo Arqueado del Hipotálamo/metabolismo , Metabolismo Energético , Riñón/metabolismo , Receptores de Leptina/metabolismo , Glándulas Suprarrenales/crecimiento & desarrollo , Proteína Relacionada con Agouti/deficiencia , Proteína Relacionada con Agouti/genética , Angiotensinógeno/deficiencia , Angiotensinógeno/genética , Animales , Núcleo Arqueado del Hipotálamo/crecimiento & desarrollo , Comunicación Autocrina , Femenino , Regulación del Desarrollo de la Expresión Génica , Riñón/crecimiento & desarrollo , Masculino , Ratones Noqueados , Miocardio/metabolismo , Comunicación Paracrina , Receptores de Leptina/deficiencia , Receptores de Leptina/genética , Albúmina Sérica/genética , Albúmina Sérica/metabolismo , Transducción de Señal
5.
J Lipid Res ; 60(12): 1983-1995, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31604805

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is considered as a liver manifestation of metabolic disorders. Previous studies indicate that the renin-angiotensin system (RAS) plays a complex role in NAFLD. As the only precursor of the RAS, decreased angiotensinogen (AGT) profoundly impacts RAS bioactivity. Here, we investigated the role of hepatocyte-derived AGT in liver steatosis. AGT floxed mice (hepAGT+/+) and hepatocyte-specific AGT-deficient mice (hepAGT-/-) were fed a Western diet and a normal laboratory diet for 12 weeks, respectively. Compared with hepAGT+/+ mice, Western diet-fed hepAGT-/- mice gained less body weight with improved insulin sensitivity. The attenuated severity of liver steatosis in hepAGT-/- mice was evidenced by histologic changes and reduced intrahepatic triglycerides. The abundance of SREBP1 and its downstream molecules, acetyl-CoA carboxylase and FASN, was suppressed in hepAGT-/- mice. Furthermore, serum derived from hepAGT+/+ mice stimulated hepatocyte SREBP1 expression, which could be diminished by protein kinase B (Akt)/mammalian target of rapamycin (mTOR) inhibition in vitro. Administration of losartan did not affect diet-induced body weight gain, liver steatosis severity, and hepatic p-Akt, p-mTOR, and SREBP1 protein abundance in hepAGT+/+ mice. These data suggest that attenuation of Western diet-induced liver steatosis in hepAGT-/- mice is associated with the alternation of the Akt/mTOR/SREBP-1c pathway.


Asunto(s)
Angiotensinógeno/metabolismo , Dieta Occidental/efectos adversos , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Enfermedad del Hígado Graso no Alcohólico/inducido químicamente , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Angiotensinógeno/deficiencia , Animales , Ácidos Grasos/metabolismo , Ratones , Enfermedad del Hígado Graso no Alcohólico/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
6.
Circulation ; 138(1): 67-79, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29371216

RESUMEN

BACKGROUND: The perivascular adipose tissue (PVAT) surrounding vessels constitutes a distinct functional integral layer of the vasculature required to preserve vascular tone under physiological conditions. However, there is little information on the relationship between PVAT and blood pressure regulation, including its potential contributions to circadian blood pressure variation. METHODS: Using unique brown adipocyte-specific aryl hydrocarbon receptor nuclear translocator-like protein 1 (Bmal1) and angiotensinogen knockout mice, we determined the vasoactivity of homogenized PVAT in aortic rings and how brown adipocyte peripheral expression of Bmal1 and angiotensinogen in PVAT regulates the amplitude of diurnal change in blood pressure in mice. RESULTS: We uncovered a peripheral clock in PVAT and demonstrated that loss of Bmal1 in PVAT reduces blood pressure in mice during the resting phase, leading to a superdipper phenotype. PVAT extracts from wild-type mice significantly induced contractility of isolated aortic rings in vitro in an endothelium-independent manner. This property was impaired in PVAT from brown adipocyte-selective Bmal1-deficient (BA-Bmal1-KO) mice. The PVAT contractile properties were mediated by local angiotensin II, operating through angiotensin II type 1 receptor-dependent signaling in the isolated vessels and linked to PVAT circadian regulation of angiotensinogen. Indeed, angiotensinogen mRNA and angiotensin II levels in PVAT of BA-Bmal1-KO mice were significantly reduced. Systemic infusion of angiotensin II, in turn, reduced Bmal1 expression in PVAT while eliminating the hypotensive phenotype during the resting phase in BA-Bmal1-KO mice. Angiotensinogen, highly expressed in PVAT, shows circadian expression in PVAT, and selective deletion of angiotensinogen in brown adipocytes recapitulates the phenotype of selective deletion of Bmal1 in brown adipocytes. Furthermore, angiotensinogen is a transcriptional target of Bmal1 in PVAT. CONCLUSIONS: These data indicate that local Bmal1 in PVAT regulates angiotensinogen expression and the ensuing increase in angiotensin II, which acts on smooth muscle cells in the vessel walls to regulate vasoactivity and blood pressure in a circadian fashion during the resting phase. These findings will contribute to a better understanding of the cardiovascular complications of circadian disorders, alterations in the circadian dipping phenotype, and cross-talk between systemic and peripheral regulation of blood pressure.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Tejido Adiposo Pardo/metabolismo , Angiotensinógeno/metabolismo , Aorta Torácica/metabolismo , Presión Sanguínea , Ritmo Circadiano , Sistema Renina-Angiotensina , Transcripción Genética , Factores de Transcripción ARNTL/deficiencia , Factores de Transcripción ARNTL/genética , Angiotensinógeno/deficiencia , Angiotensinógeno/genética , Animales , Presión Sanguínea/genética , Ritmo Circadiano/genética , Genotipo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Sistema Renina-Angiotensina/genética , Descanso , Transducción de Señal , Factores de Tiempo , Vasoconstricción
7.
Physiol Rep ; 4(1)2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26755736

RESUMEN

The role of intranephron angiotensinogen (AGT) in blood pressure (BP) regulation is not fully understood. Previous studies showed that proximal tubule-specific overexpression of AGT increases BP, whereas proximal tubule-specific deletion of AGT did not alter BP. The latter study may not have completely eliminated nephron AGT production; in addition, BP was only assessed on a normal salt diet. To evaluate this issue in greater detail, we developed mice with inducible nephron-wide AGT deletion. Mice were generated which were hemizygous for the Pax8-rtTA and LC-1 transgenes and homozygous for loxP-flanked AGT alleles to achieve nephron-wide AGT disruption after doxycycline induction. Compared to controls, AGT knockout (KO) mice demonstrated markedly reduced renal AGT immunostaining, mRNA, and protein levels; unexpectedly AGT KO mice had reduced AGT mRNA levels in the liver along with 50% reduction in plasma AGT levels. BP was significantly lower in the AGT KO mice compared to controls fed a normal, low, or high Na(+) intake, with the highest BP reduction on a low Na(+) diet. Regardless of Na(+) intake, AGT KO mice had higher plasma renin concentration (PRC) and markedly reduced urinary AGT levels compared to controls. Following angiotensin-II (Ang-II) infusion, AGT KO mice demonstrated an attenuated hypertensive response despite similar suppression of PRC in the two groups. Taken together, these data suggest that nephron-derived AGT may be involved in Ang-II-dependent hypertension, however, a clear role for nephron-derived AGT in physiological BP regulation remains to be determined.


Asunto(s)
Angiotensina II/toxicidad , Angiotensinógeno/deficiencia , Hipertensión/inducido químicamente , Hipertensión/metabolismo , Nefronas/fisiología , Sodio en la Dieta/efectos adversos , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
8.
Arterioscler Thromb Vasc Biol ; 36(2): 256-65, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26681751

RESUMEN

OBJECTIVE: This study determined whether angiotensinogen (AGT) has angiotensin II-independent effects using multiple genetic and pharmacological manipulations. APPROACH AND RESULTS: All study mice were in low-density lipoprotein receptor -/- background and fed a saturated fat-enriched diet. In mice with floxed alleles and a neomycin cassette in intron 2 of the AGT gene (hypoAGT mice), plasma AGT concentrations were >90% lower compared with their wild-type littermates. HypoAGT mice had lower systolic blood pressure, less atherosclerosis, and diminished body weight gain and liver steatosis. Low plasma AGT concentrations and all phenotypes were recapitulated in mice with hepatocyte-specific deficiency of AGT or pharmacological inhibition of AGT by antisense oligonucleotide administration. In contrast, inhibition of AGT cleavage by a renin inhibitor, aliskiren, failed to alter body weight gain and liver steatosis in low-density lipoprotein receptor -/- mice. In mice with established adiposity, administration of AGT antisense oligonucleotide versus aliskiren led to equivalent reductions of systolic blood pressure and atherosclerosis. AGT antisense oligonucleotide administration ceased body weight gain and further reduced body weight, whereas aliskiren did not affect body weight gain during continuous saturated fat-enriched diet feeding. Structural comparisons of AGT proteins in zebrafish, mouse, rat, and human revealed 4 highly conserved sequences within the des(angiotensin I)AGT domain. des(angiotensin I)AGT, through adeno-associated viral infection in hepatocyte-specific AGT-deficient mice, increased body weight gain and liver steatosis, but did not affect atherosclerosis. CONCLUSIONS: AGT contributes to body weight gain and liver steatosis through functions of the des(angiotensin I)AGT domain, which are independent of angiotensin II production.


Asunto(s)
Angiotensina II/metabolismo , Angiotensinógeno/metabolismo , Aterosclerosis/metabolismo , Hígado Graso/metabolismo , Hepatocitos/metabolismo , Hipertensión/metabolismo , Hígado/metabolismo , Amidas/farmacología , Secuencia de Aminoácidos , Angiotensinógeno/deficiencia , Angiotensinógeno/genética , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/prevención & control , Presión Sanguínea , Secuencia Conservada , Dependovirus/genética , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Hígado Graso/genética , Hígado Graso/patología , Hígado Graso/prevención & control , Fumaratos/farmacología , Vectores Genéticos , Genotipo , Hepatocitos/patología , Hipertensión/genética , Hipertensión/fisiopatología , Hipertensión/prevención & control , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Moleculares , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Fenotipo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Receptores de LDL/deficiencia , Receptores de LDL/genética , Renina/antagonistas & inhibidores , Renina/metabolismo , Transducción de Señal , Factores de Tiempo , Transducción Genética , Aumento de Peso
9.
Hypertension ; 66(4): 836-42, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26303292

RESUMEN

We recently demonstrated that adipocyte deficiency of angiotensinogen (AGT) ablated high-fat diet-induced elevations in plasma angiotensin II (Ang II) concentrations and obesity-hypertension in male mice. Hepatocytes are the predominant source of systemic AGT. Therefore, in this study, we defined the contribution of hepatocyte-derived AGT to obesity-induced elevations in plasma AGT concentrations and hypertension. Male Agt(fl/fl) mice expressing albumin-driven Cre recombinase were bred to female Agt(fl/fl) mice to generate Agt(fl/fl) or hepatocyte AGT-deficient male mice (Agt(Alb)). Mice were fed a low-fat or high-fat diet for 16 weeks. Hepatocyte AGT deficiency had no significant effect on body weight. Plasma AGT concentrations were increased in obese Agt(fl/fl) mice. Hepatocyte AGT deficiency markedly reduced plasma AGT and Ang II concentrations in lean and obese mice. Moreover, hepatocyte AGT deficiency reduced the content and release of AGT from adipose explants. Systolic blood pressure was markedly decreased in lean (by 18 mm Hg) and obese Agt(Alb) mice (by 54 mm Hg) compared with Agt(fl/fl) controls. To define mechanisms, we quantified effects of Ang II on mRNA abundance of megalin, an AGT uptake transporter, in 3T3-L1 adipocytes. Ang II stimulated adipocyte megalin mRNA abundance and decreased media AGT concentrations. These results demonstrate that hepatocytes are the predominant source of systemic AGT in both lean and obese mice. Moreover, reductions in plasma angiotensin concentrations in obese hepatocyte AGT-deficient mice may have limited megalin-dependent uptake of AGT into adipocytes for the production of Ang II in the development of obesity-hypertension.


Asunto(s)
Angiotensinógeno/genética , Presión Sanguínea , Regulación de la Expresión Génica , Hepatocitos/metabolismo , Hipertensión/metabolismo , Obesidad/metabolismo , Delgadez/metabolismo , Angiotensinógeno/deficiencia , Animales , Peso Corporal , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Hepatocitos/patología , Hipertensión/genética , Hipertensión/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/complicaciones , Obesidad/fisiopatología , ARN Mensajero/genética , Delgadez/complicaciones , Delgadez/fisiopatología
10.
Kidney Int ; 85(5): 1068-77, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24284520

RESUMEN

Intrarenal angiotensin II is increased in kidney diseases independently of plasma angiotensin II and is thought to promote progressive deterioration of renal architecture. Here we investigated the mechanism of enhanced renal angiotensin II generation in kidney glomerular diseases. For this, kidney- or liver-specific angiotensinogen gene (Agt) knockout was superimposed on the mouse model of inducible podocyte injury (NEP25). Seven days after induction of podocyte injury, renal angiotensin II was increased ninefold in NEP25 mice with intact Agt, accompanied by increases in urinary albumin and angiotensinogen excretion, renal angiotensinogen protein, and its mRNA. Kidney Agt knockout attenuated renal Agt mRNA but not renal angiotensin II, renal, or urinary angiotensinogen protein. In contrast, liver Agt knockout markedly reduced renal angiotensin II to 18.7% of that of control NEP25 mice, renal and urinary angiotensinogen protein, but not renal Agt mRNA. Renal angiotensin II had no relationship with renal Agt mRNA, or with renal renin mRNA, which was elevated in liver Agt knockouts. Kidney and liver dual Agt knockout mice showed phenotypes comparable to those of liver Agt knockout mice. Thus, increased renal angiotensin II generation upon severe podocyte injury is attributed to increased filtered angiotensinogen of liver origin resulting from loss of macromolecular barrier function of the glomerular capillary wall that occurs upon severe podocyte injury.


Asunto(s)
Angiotensina II/metabolismo , Angiotensinógeno/metabolismo , Enfermedades Renales/metabolismo , Túbulos Renales Proximales/metabolismo , Hígado/metabolismo , Podocitos/metabolismo , Albuminuria/genética , Albuminuria/metabolismo , Albuminuria/patología , Angiotensinógeno/deficiencia , Angiotensinógeno/genética , Angiotensinógeno/orina , Animales , Anticuerpos Monoclonales , Modelos Animales de Enfermedad , Exotoxinas , Genotipo , Enfermedades Renales/inducido químicamente , Enfermedades Renales/genética , Enfermedades Renales/patología , Túbulos Renales Proximales/patología , Proteínas de la Membrana/metabolismo , Ratones Noqueados , Fenotipo , Podocitos/patología , ARN Mensajero/metabolismo , Sodio/metabolismo , Factores de Tiempo , Regulación hacia Arriba
11.
J Am Soc Nephrol ; 23(7): 1181-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22518004

RESUMEN

Angiotensin II content in the kidney is much higher than in the plasma, and it increases more in kidney diseases through an uncertain mechanism. Because the kidney abundantly expresses angiotensinogen mRNA, transcriptional dysregulation of angiotensinogen within the kidney is one potential cause of increased renal angiotensin II in the setting of disease. Here, we observed that kidney-specific angiotensinogen knockout mice had levels of renal angiotensinogen protein and angiotensin II that were similar to those levels of control mice. In contrast, liver-specific knockout of angiotensinogen nearly abolished plasma and renal angiotensinogen protein and renal tissue angiotensin II. Immunohistochemical analysis in mosaic proximal tubules of megalin knockout mice revealed that angiotensinogen protein was incorporated selectively in megalin-intact cells of the proximal tubule, indicating that the proximal tubule reabsorbs filtered angiotensinogen through megalin. Disruption of the filtration barrier in a transgenic mouse model of podocyte-selective injury increased renal angiotensin II content and markedly increased both tubular and urinary angiotensinogen protein without an increase in renal renin activity, supporting the dependency of renal angiotensin II generation on filtered angiotensinogen. Taken together, these data suggest that liver-derived angiotensinogen is the primary source of renal angiotensinogen protein and angiotensin II. Furthermore, an abnormal increase in the permeability of the glomerular capillary wall to angiotensinogen, which characterizes proteinuric kidney diseases, enhances the synthesis of renal angiotensin II.


Asunto(s)
Angiotensina II/metabolismo , Angiotensinógeno/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Angiotensina II/deficiencia , Angiotensina II/genética , Angiotensinógeno/deficiencia , Angiotensinógeno/genética , Animales , Riñón/patología , Hígado/patología , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/deficiencia , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Animales , Fenotipo , ARN Mensajero/metabolismo
12.
Hypertension ; 59(6): 1188-94, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22526255

RESUMEN

The (pro)renin receptor is a newly discovered member of the brain renin-angiotensin system. To investigate the role of brain (pro)renin receptor in hypertension, adeno-associated virus-mediated (pro)renin receptor short hairpin RNA was used to knockdown (pro)renin receptor expression in the brain of nontransgenic normotensive and human renin-angiotensinogen double-transgenic hypertensive mice. Blood pressure was monitored using implanted telemetric probes in conscious animals. Real-time PCR and immunostaining were performed to determine (pro)renin receptor, angiotensin II type 1 receptor, and vasopressin mRNA levels. Plasma vasopressin levels were determined by ELISA. Double-transgenic mice exhibited higher blood pressure, elevated cardiac and vascular sympathetic tone, and impaired spontaneous baroreflex sensitivity. Intracerebroventricular delivery of (pro)renin receptor short-hairpin RNA significantly reduced blood pressure, cardiac and vasomotor sympathetic tone, and improved baroreflex sensitivity compared with the control virus treatment in double-transgenic mice. (Pro)renin receptor knockdown significantly reduced angiotensin II type 1 receptor and vasopressin levels in double-transgenic mice. These data indicate that (pro)renin receptor knockdown in the brain attenuates angiotensin II-dependent hypertension and is associated with a decrease in sympathetic tone and an improvement of the baroreflex sensitivity. In addition, brain-targeted (pro)renin receptor knockdown is associated with downregulation of angiotensin II type 1 receptor and vasopressin levels. We conclude that central (pro)renin receptor contributes to the pathogenesis of hypertension in human renin-angiotensinogen transgenic mice.


Asunto(s)
Angiotensina II/metabolismo , Encéfalo/metabolismo , Hipertensión/metabolismo , Receptores de Superficie Celular/metabolismo , Angiotensina II/sangre , Angiotensinógeno/deficiencia , Angiotensinógeno/genética , Animales , Barorreflejo/genética , Barorreflejo/fisiología , Presión Sanguínea/genética , Presión Sanguínea/fisiología , Western Blotting , Línea Celular Tumoral , Humanos , Hipertensión/genética , Hipertensión/fisiopatología , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Interferencia de ARN , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Receptores de Superficie Celular/genética , Renina/deficiencia , Renina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Vasopresinas/sangre , Vasopresinas/genética , Vasopresinas/metabolismo , Receptor de Prorenina
13.
Brain Res ; 1448: 101-10, 2012 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-22405726

RESUMEN

Previous evidence indicates that a balance between inhibitory gabaergic and excitatory angiotensinergic factors in the PVN is important for cardiovascular control. We investigated the cardiovascular response evoked from activation or blockade of GABA(A) receptors in the paraventricular nucleus (PVN), in transgenic rats with low brain angiotensinogen [TGR(ASrAOGEN)]. Brain Ang II and Ang-(1-7) levels were also determined. In functional experiments, TGR(ASrAOGEN) and Sprague-Dawley rats (SD, control) were anesthetized with urethane and blood pressure (BP), heart rate (HR) and renal sympathetic nerve activity (RSNA) were recorded. Brain Ang II and Ang-(1-7) levels were largely reduced in TGR(ASrAOGEN) compared with SD rats. Inhibition of PVN neurons with the GABA(A) agonist, muscimol (1 nmol/100 nL), resulted in an attenuated fall in all cardiovascular variables in TGR(ASrAOGEN) compared with SD rats. This difference was particularly pronounced in HR (TGR Mus -23±6 bpm vs. -77±9 bpm SD Mus; P<0.05) and RSNA (TGR -3±10% vs.-29±8% SD; P<0.05). Furthermore, the sympathetic response evoked by blockade of GABA(A) receptors in the PVN of TGR(ASrAOGEN) was also largely suppressed. The present data indicate that the sympathetic outflow mediated by PVN neurons under basal conditions is suppressed in TGR(ASrAOGEN) rats corroborating the functional significance of brain angiotensin production in the central regulation of sympathetic output to the cardiovascular system.


Asunto(s)
Angiotensinógeno/deficiencia , Química Encefálica/efectos de los fármacos , Agonistas del GABA/farmacología , Antagonistas del GABA/farmacología , Hemodinámica/efectos de los fármacos , Hemodinámica/fisiología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Receptores de GABA-A/fisiología , Animales , Antihipertensivos/farmacología , Presión Sanguínea/efectos de los fármacos , Química Encefálica/genética , Expresión Génica/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Compuestos de Hexametonio/farmacología , Riñón/inervación , Riñón/fisiología , Radioinmunoensayo , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Sistema Nervioso Simpático/fisiología , Transgenes/genética
14.
Pediatr Res ; 67(6): 573-8, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20496471

RESUMEN

Mutations in the genes encoding components of the renin-angiotensin system (RAS) in mice or humans cause congenital abnormalities of the kidney and urinary tract. We hypothesized that absence of angiotensin (Ang) II in angiotensinogen (AGT)-deficient mice leads to defects in ureteric bud (UB) branching and that RAS genes are critically dependent on histone deacetylase (HDAC) activity. The number of UB tips was lower in AGT-/- compared with AGT+/+ embryonic (E) day E13.5 metanephroi (24+/-1.5 versus 36+/-3.7, p<0.05). Real-time RT-PCR demonstrated that pharmacological inhibition of HDAC activity with Scriptaid increases AGT, renin, angiotensin-converting enzyme (ACE), and AT1 receptor (AT1R) mRNA levels in E12.5 mouse metanephroi and early mesenchymal (MK3) cells. Furthermore, Scriptaid enhanced Ang II induced decrease in Sprouty (Spry) 1 gene expression in cultured UB cells. Treatment of intact E12.5 mouse metanephroi grown ex vivo with Ang II (10(-5) M, 24 h) increased HDAC-1 and decreased total acetylated histone H3 protein levels. These findings indicate that lack of endogenous Ang II in AGT-deficient mice inhibits UB branching. We conclude that intact RAS is critical in structural integrity of the renal collecting system and that UB morphogenetic program genes, such as AGT, renin, ACE, AT1R, or Spry1, are epigenetically controlled via HDACs.


Asunto(s)
Angiotensina II/metabolismo , Histona Desacetilasa 1/metabolismo , Sistema Renina-Angiotensina , Uréter/enzimología , Acetilación , Proteínas Adaptadoras Transductoras de Señales , Angiotensina II/genética , Angiotensinógeno/deficiencia , Angiotensinógeno/genética , Animales , Células Cultivadas , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Edad Gestacional , Histona Desacetilasa 1/antagonistas & inhibidores , Histona Desacetilasa 1/genética , Inhibidores de Histona Desacetilasas/farmacología , Histonas/metabolismo , Hidroxilaminas/farmacología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfogénesis , Fosfoproteínas/metabolismo , Quinolinas/farmacología , ARN Mensajero/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/genética , Técnicas de Cultivo de Tejidos , Uréter/efectos de los fármacos , Uréter/embriología
15.
J Endocrinol ; 204(1): 85-92, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19808775

RESUMEN

AT(1) blockers attenuate hypothalamo-pituitary-adrenal (HPA) axis reactivity in hypertension independently of their potency to lower blood pressure. A reduced pituitary sensitivity to CRH and a downregulation of hypothalamic CRH expression have been suggested to influence HPA axis activity during chronic AT(1) blockade. This study was aimed at confirming the role of central angiotensin II in regulating HPA reactivity by using the transgenic rat TGR(ASrAOGEN), a model featuring low levels of brain angiotensinogen. Different stress tests were performed to determine HPA reactivity in TGR(ASrAOGEN) and appropriate controls. In TGR(ASrAOGEN), blood pressure was diminished compared to controls. The corticosterone response to a CRH or ACTH challenge and a forced swim test was more distinct in TGR(ASrAOGEN) than it was in controls and occurred independently of a concurrent enhancement in ACTH. Using quantitative real-time PCR, we found increased mRNA levels of melanocortin 2 (Mc2r) and AT(2) receptors (Agtr2) in the adrenals of TGR(ASrAOGEN), whereas mRNA levels of Crh, Pomc, and AT(1) receptors (Agtr1) remained unchanged in hypothalami and pituitary glands. Since stress responses were increased rather than attenuated in TGR(ASrAOGEN), we conclude that the reduced HPA reactivity during AT(1) blockade could not be mimicked in a specific transgenic rat model featuring a centrally inactivated renin-angiotensin-aldosterone system. The ACTH independency of the enhanced corticosterone release during CRH test and the enhanced corticosterone response to ACTH rather indicates an adrenal mechanism. The upregulation of adrenal MC2 and AT(2) receptors seems to be involved in the stimulated facilitation of adrenal corticosterone release for effectuating the stimulated stress responses.


Asunto(s)
Angiotensina II/metabolismo , Angiotensinógeno/deficiencia , Encéfalo/metabolismo , Sistema Hipotálamo-Hipofisario/fisiología , Receptor de Angiotensina Tipo 2/metabolismo , Receptor de Melanocortina Tipo 2/metabolismo , Estrés Fisiológico/fisiología , Glándulas Suprarrenales/metabolismo , Hormona Adrenocorticotrópica/farmacología , Animales , Presión Sanguínea/fisiología , Corticosterona/metabolismo , Hormona Liberadora de Corticotropina/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Receptor de Angiotensina Tipo 2/genética , Receptor de Melanocortina Tipo 2/genética , Sistema Renina-Angiotensina/fisiología , Natación , Regulación hacia Arriba
16.
Neurosci Lett ; 436(2): 163-6, 2008 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-18384957

RESUMEN

The neuropeptide galanin is elevated in the cardiac sympathetic innervation after myocardial infarction (MI). Galanin inhibits vagal transmission and may support the regeneration of sympathetic nerves, thereby contributing to the development of arrhythmia and sudden cardiac death after MI. The reason for increased galanin production in sympathetic neurons after myocardial infarction is not known. Cardiac sympathetic neurons are activated chronically after cardiac ischemia-reperfusion, and activation of sympathetic neurons in culture stimulates galanin expression. Therefore, we tested the hypothesis that increased sympathetic nerve activity stimulates galanin expression in cardiac sympathetic neurons after myocardial infarction. To test this hypothesis we used TGR(ASrAOGEN) transgenic rats, which lack brain angiotensinogen and do not exhibit post-infarct sympathetic hyperactivity. Hearts and stellate ganglia were collected 1 week after ischemia-reperfusion. Galanin mRNA was quantified by real-time PCR and peptide content was assayed by enzyme-linked immunosorbent assay. Galanin mRNA increased approximately 3-fold after MI in cardiac sympathetic neurons of both genotypes compared to unoperated and sham controls. Left ventricular galanin content, however, increased after MI only in Sprague-Dawley rats and not in AOGEN rats. These data suggest that post-infarct cardiac sympathetic hyperactivity stimulates galanin peptide production but is not required for increased galanin mRNA expression.


Asunto(s)
Galanina/metabolismo , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Sistema Nervioso Simpático/fisiología , Angiotensinógeno/deficiencia , Animales , Animales Modificados Genéticamente , Estudios de Casos y Controles , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Galanina/genética , Regulación de la Expresión Génica/fisiología , Masculino , Reperfusión Miocárdica/métodos , Ratas , Ratas Sprague-Dawley , Ganglio Estrellado/metabolismo
17.
J Neuroendocrinol ; 20(2): 182-7, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18047554

RESUMEN

In transgenic rats, TGR(ASrAOGEN)680, with reduced glial expression of angiotensinogen, changes in brain angiotensinogen are associated with reductions in serotonin (5-HT) content and/or 5-HT metabolism as determined in various brain regions, including the hypothalamus. These rats showed an anxious phenotype upon a first behavioural screen. The present study aimed to extend the search for functional consequences of changes in brain 5-HT with respect to feeding behaviour in these transgenic rats. In feeding experiments, rats were treated with the anorectic drug fenfluramine to probe for functional changes in the serotonergic satiety system. Fenfluramine (0.3 mg/kg, i.p.) reduced food intake in TGR(ASrAOGEN)680 rats whereas the minimal effective dose in wild-type rats was 3 mg/kg, i.p. Although, in the cortex, no differences were apparent in the expression of serotonin 5-HT(1A), 5-HT(1B), 5-HT(2C) receptor and 5-HT transporter mRNAs between TGR(ASrAOGEN)680 and wild-type rats, the expression of mRNAs for the 5-HT(2C) receptor and 5-HT transporter mRNA were significantly higher in the hypothalamus of TGR(ASrAOGEN)680 rats compared to wild-type rats. No differences were found in the mRNA levels for hypothalamic 5-HT(1A) and 5-HT(1B) receptors between TGR(ASrAOGEN)680 and wild-type rats. Taken together, these findings suggest that the transgenic effect on the brain 5-HT system is paralleled by functional changes of the serotonergic feeding system.


Asunto(s)
Angiotensinógeno/deficiencia , Encéfalo/fisiología , Respuesta de Saciedad/fisiología , Serotonina/fisiología , Animales , Animales Modificados Genéticamente , Peso Corporal , Encéfalo/metabolismo , Corteza Cerebral/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Conducta Alimentaria/fisiología , Fenfluramina/farmacología , Hipotálamo/metabolismo , Masculino , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Serotonina/genética , Respuesta de Saciedad/efectos de los fármacos , Serotonina/farmacología , Serotoninérgicos/farmacología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética
18.
Am J Physiol Heart Circ Physiol ; 294(1): H99-H106, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17951370

RESUMEN

The balance between norepinephrine (NE) synthesis, release, and reuptake is disrupted after acute myocardial infarction, resulting in elevated extracellular NE. Stimulation of sympathetic neurons in vitro increases NE synthesis and the synthetic enzyme tyrosine hydroxylase (TH) to a greater extent than it increases NE reuptake and the NE transporter (NET), which removes NE from the extracellular space. We used TGR(ASrAOGEN) transgenic rats, which lack postinfarct sympathetic hyperactivity, to test the hypothesis that increased cardiac sympathetic nerve activity accounts for the imbalance in TH and NET expression in these neurons after myocardial infarction. TH and NET mRNA levels were identical in the stellate ganglia of unoperated TGR(ASrAOGEN) rats compared with Sprague Dawley (SD) controls, but the threefold increase in TH and twofold increase in NET mRNA seen in the stellate ganglia of SD rats 1 wk after ischemia-reperfusion was absent in TGR(ASrAOGEN) rats. Similarly, the increase in TH and NET protein observed in the base of the SD ventricle was absent in the base of the TGR (ASrAOGEN) ventricle. Neuronal TH content was depleted in the left ventricle of both genotypes, whereas NET was unchanged. Basal heart rate and cardiac function were similar in both genotypes, but TGR(ASrAOGEN) hearts were more sensitive to the beta-agonist dobutamine. Tyramine-induced release of endogenous NE generated similar changes in ventricular pressure and contractility in both genotypes, but postinfarct relaxation was enhanced in TGR(ASrAOGEN) hearts. These data support the hypothesis that postinfarct sympathetic hyperactivity is the major stimulus increasing TH and NET expression in cardiac neurons.


Asunto(s)
Corazón/inervación , Infarto del Miocardio/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Sistema Nervioso Simpático/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Agonistas Adrenérgicos beta/farmacología , Angiotensinógeno/deficiencia , Angiotensinógeno/genética , Angiotensinógeno/metabolismo , Animales , Animales Modificados Genéticamente , Vasos Coronarios/cirugía , Modelos Animales de Enfermedad , Dobutamina/farmacología , Femenino , Corazón/efectos de los fármacos , Frecuencia Cardíaca , Ligadura , Masculino , Contracción Miocárdica , Infarto del Miocardio/enzimología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/fisiopatología , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Ganglio Estrellado/enzimología , Ganglio Estrellado/metabolismo , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/enzimología , Sistema Nervioso Simpático/fisiopatología , Simpatomiméticos/farmacología , Tiramina/farmacología , Tirosina 3-Monooxigenasa/genética , Regulación hacia Arriba , Función Ventricular Izquierda
19.
J Neurosci ; 27(34): 9032-42, 2007 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-17715340

RESUMEN

The blood-brain barrier (BBB) restricts molecular and cellular trafficking between the blood and the CNS. Although astrocytes are known to control BBB permeability, the molecular determinants of this effect remain unknown. We show that angiotensinogen (AGT) produced and secreted by astrocytes is cleaved into angiotensin II (AngII) and acts on type 1 angiotensin receptors (AT1) expressed by BBB endothelial cells (ECs). Activation of AT1 restricts the passage of molecular tracers across human BBB-derived ECs through threonine-phosphorylation of the tight junction protein occludin and its mobilization to lipid raft membrane microdomains. We also show that AGT knock-out animals have disorganized occludin strands at the level of the BBB and a diffuse accumulation of the endogenous serum protein plasminogen in the CNS, compared with wild-type animals. Finally, we demonstrate a reduction in the number of AGT-immunopositive perivascular astrocytes in multiple sclerosis (MS) lesions, which correlates with a reduced expression of occludin similarly seen in the CNS of AGT knock-out animals. Such a reduction in astrocyte-expressed AGT and AngII is dependent, in vitro, on the proinflammatory cytokines tumor necrosis factor-alpha and interferon-gamma. Our study defines a novel physiological role for AngII in the CNS and suggests that inflammation-induced downregulation of AngII production by astrocytes is involved in BBB dysfunction in MS lesions.


Asunto(s)
Angiotensina II/farmacología , Barrera Hematoencefálica/citología , Células Endoteliales/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Esclerosis Múltiple/metabolismo , Adulto , Angiotensinógeno/deficiencia , Angiotensinógeno/metabolismo , Animales , Astrocitos/química , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Medios de Cultivo Condicionados/farmacología , Citocinas/genética , Citocinas/metabolismo , Feto , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Humanos , Masculino , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Persona de Mediana Edad , Esclerosis Múltiple/patología , Ocludina , Receptores de Angiotensina/metabolismo , Proteínas S100/metabolismo
20.
Circ Res ; 99(10): 1125-31, 2006 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-17053195

RESUMEN

The subfornical organ (SFO) of the brain has long been considered a critical integrating center for the cardiovascular actions of the renin-angiotensin system (RAS). Early reports of angiotensin II (Ang II) immunoreactivity in the SFO and its neural projections to downstream cardiovascular nuclei raised the possibility that Ang II is produced locally and functions as a putative neurotransmitter in these circuits. However, evidence of functionally significant de novo synthesis of Ang II in the SFO has been lacking. Here, implementing spatiotemporally restricted gene ablation by way of the Cre recombinase/loxP system, we provide the first direct evidence that the local RAS in the SFO has a critical role in blood pressure regulation. Using a transgenic mouse harboring an angiotensinogen (AGT) gene modified for Cre-mediated deletion (hAGT(flox)), in combination with gene transfer of an adenovirus encoding Cre targeted to the SFO, we show that deletion of the Ang II substrate in this brain region nearly abolishes the pressor and bradycardic effects of renin infused in the CNS. Immunohistochemical analyses verified intense and restricted expression of Cre in the SFO, which paralleled the decrease in AGT expression selectively in this site. Further physiological studies confirmed the integrity of central angiotensinergic and nonangiotensinergic cardiovascular response systems in the Cre-treated mice. In addition to establishing that AGT expression in the SFO and its local conversion to Ang II has a profound effect on blood pressure, this study provides proof-of-principle of the utility of this approach for dissecting the brain RAS and other complex systems in CNS cardiovascular circuits.


Asunto(s)
Angiotensinógeno/genética , Presión Sanguínea/fisiología , Órgano Subfornical/fisiología , Adenoviridae/genética , Angiotensina II/biosíntesis , Angiotensinógeno/sangre , Angiotensinógeno/deficiencia , Angiotensinógeno/metabolismo , Animales , Femenino , Vectores Genéticos/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Recombinasas/genética , Recombinasas/metabolismo , Renina/antagonistas & inhibidores , Renina/farmacología , Órgano Subfornical/efectos de los fármacos , Órgano Subfornical/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA