Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Pharmacol Rev ; 74(3): 462-505, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35710133

RESUMEN

The concept of local formation of angiotensin II in the kidney has changed over the last 10-15 years. Local synthesis of angiotensinogen in the proximal tubule has been proposed, combined with prorenin synthesis in the collecting duct. Binding of prorenin via the so-called (pro)renin receptor has been introduced, as well as megalin-mediated uptake of filtered plasma-derived renin-angiotensin system (RAS) components. Moreover, angiotensin metabolites other than angiotensin II [notably angiotensin-(1-7)] exist, and angiotensins exert their effects via three different receptors, of which angiotensin II type 2 and Mas receptors are considered renoprotective, possibly in a sex-specific manner, whereas angiotensin II type 1 (AT1) receptors are believed to be deleterious. Additionally, internalized angiotensin II may stimulate intracellular receptors. Angiotensin-converting enzyme 2 (ACE2) not only generates angiotensin-(1-7) but also acts as coronavirus receptor. Multiple, if not all, cardiovascular diseases involve the kidney RAS, with renal AT1 receptors often being claimed to exert a crucial role. Urinary RAS component levels, depending on filtration, reabsorption, and local release, are believed to reflect renal RAS activity. Finally, both existing drugs (RAS inhibitors, cyclooxygenase inhibitors) and novel drugs (angiotensin receptor/neprilysin inhibitors, sodium-glucose cotransporter-2 inhibitors, soluble ACE2) affect renal angiotensin formation, thereby displaying cardiovascular efficacy. Particular in the case of the latter three, an important question is to what degree they induce renoprotection (e.g., in a renal RAS-dependent manner). This review provides a unifying view, explaining not only how kidney angiotensin formation occurs and how it is affected by drugs but also why drugs are renoprotective when altering the renal RAS. SIGNIFICANCE STATEMENT: Angiotensin formation in the kidney is widely accepted but little understood, and multiple, often contrasting concepts have been put forward over the last two decades. This paper offers a unifying view, simultaneously explaining how existing and novel drugs exert renoprotection by interfering with kidney angiotensin formation.


Asunto(s)
Angiotensinógeno , Enfermedades Cardiovasculares , Femenino , Humanos , Masculino , Angiotensina II/metabolismo , Enzima Convertidora de Angiotensina 2 , Angiotensinógeno/metabolismo , Enfermedades Cardiovasculares/metabolismo , Sistemas de Liberación de Medicamentos , Riñón/irrigación sanguínea , Riñón/metabolismo , Renina/metabolismo , Sistema Renina-Angiotensina , Inhibidores del Cotransportador de Sodio-Glucosa 2/metabolismo
2.
Int J Mol Sci ; 24(8)2023 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-37108577

RESUMEN

It has been our pleasure to have been able to develop two special issues within the International Journal of Molecular Sciences: (1) Renin-Angiotensin-Aldosterone System in Pathologies and (2) Renin-Angiotensin-Aldosterone System in Metabolism & Disease [...].


Asunto(s)
Enfermedades Metabólicas , Sistema Renina-Angiotensina , Humanos , Aldosterona , Renina/metabolismo , Angiotensina II/metabolismo
3.
Int J Mol Sci ; 23(5)2022 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-35269547

RESUMEN

Contrary to public perception, hypertension remains one of the most important public health problems in the United States, affecting 46% of adults with increased risk for heart attack, stroke, and kidney diseases. The mechanisms underlying poorly controlled hypertension remain incompletely understood. Recent development in the Cre/LoxP approach to study gain or loss of function of a particular gene has significantly helped advance our new insights into the role of proximal tubule angiotensin II (Ang II) and its AT1 (AT1a) receptors in basal blood pressure control and the development of Ang II-induced hypertension. This novel approach has provided us and others with an important tool to generate novel mouse models with proximal tubule-specific loss (deletion) or gain of the function (overexpression). The objective of this invited review article is to review and discuss recent findings using novel genetically modifying proximal tubule-specific mouse models. These new studies have consistently demonstrated that deletion of AT1 (AT1a) receptors or its direct downstream target Na+/H+ exchanger 3 (NHE3) selectively in the proximal tubules of the kidney lowers basal blood pressure, increases the pressure-natriuresis response, and induces natriuretic responses, whereas overexpression of an intracellular Ang II fusion protein or AT1 (AT1a) receptors selectively in the proximal tubules increases proximal tubule Na+ reabsorption, impairs the pressure-natriuresis response, and elevates blood pressure. Furthermore, the development of Ang II-induced hypertension by systemic Ang II infusion or by proximal tubule-specific overexpression of an intracellular Ang II fusion protein was attenuated in mutant mice with proximal tubule-specific deletion of AT1 (AT1a) receptors or NHE3. Thus, these recent studies provide evidence for and new insights into the important roles of intratubular Ang II via AT1 (AT1a) receptors and NHE3 in the proximal tubules in maintaining basal blood pressure homeostasis and the development of Ang II-induced hypertension.


Asunto(s)
Angiotensina II/metabolismo , Hipertensión/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Angiotensina II/genética , Animales , Presión Sanguínea , Modelos Animales de Enfermedad , Mutación con Ganancia de Función , Humanos , Hipertensión/genética , Mutación con Pérdida de Función , Ratones , Receptor de Angiotensina Tipo 1/genética , Intercambiador 3 de Sodio-Hidrógeno/metabolismo
4.
Clin Sci (Lond) ; 135(15): 1825-1843, 2021 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-34282828

RESUMEN

In the present study, we tested the hypothesis that there are significant sex differences in angiotensin II (Ang II)-induced hypertension and kidney injury using male and female wildtype (WT) and proximal tubule-specific AT1a receptor knockout mice (PT-Agtr1a-/-). Twelve groups (n=8-12 per group) of adult male and female WT and PT-Agtr1a-/- mice were infused with a pressor dose of Ang II via osmotic minipump for 2 weeks (1.5 mg/kg/day, i.p.) and simultaneously treated with or without losartan (20 mg/kg/day, p.o.) to determine the respective roles of AT1a receptors in the proximal tubules versus systemic tissues. Basal systolic, diastolic, and mean arterial pressure were approximately 13 ± 3 mmHg lower (P<0.01), while basal 24-h urinary Na+, K+, and Cl- excretion were significantly higher in both male and female PT-Agtr1a-/- mice than WT controls (P<0.01) without significant sex differences between different strains. Both male and female WT and PT-Agtr1a-/- mice developed hypertension (P<0.01), and the magnitudes of the pressor responses to Ang II were similar between male and female WT and PT-Agtr1a-/- mice (n.s.). Likewise, Ang II-induced hypertension was significantly attenuated in both male and female PT-Agtr1a-/- mice (P<0.01). Furthermore, losartan attenuated the hypertensive responses to Ang II to similar extents in both male and female WT and PT-Agtr1a-/- mice. Finally, Ang II-induced kidney injury was attenuated in PT-Agtr1a-/- mice (P<0.01). In conclusion, the present study demonstrates that deletion of AT1a receptors in the proximal tubules of the kidney attenuates Ang II-induced hypertension and kidney injury without revealing significant sex differences.


Asunto(s)
Presión Arterial , Hipertensión/metabolismo , Enfermedades Renales/metabolismo , Túbulos Renales Proximales/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Sistema Renina-Angiotensina , Angiotensina II , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Antihipertensivos/farmacología , Presión Arterial/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Fibrosis , Hipertensión/inducido químicamente , Hipertensión/fisiopatología , Hipertensión/prevención & control , Enfermedades Renales/inducido químicamente , Enfermedades Renales/fisiopatología , Enfermedades Renales/prevención & control , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/fisiopatología , Túbulos Renales Proximales/ultraestructura , Losartán/farmacología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Angiotensina Tipo 1/genética , Sistema Renina-Angiotensina/efectos de los fármacos , Caracteres Sexuales , Factores Sexuales , Transducción de Señal
5.
Physiol Genomics ; 51(4): 97-108, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30849009

RESUMEN

The sodium (Na+)/hydrogen (H+) exchanger 3 (NHE3) and sodium-potassium adenosine triphosphatase (Na+/K+-ATPase) are two of the most important Na+ transporters in the proximal tubules of the kidney. On the apical membrane side, NHE3 primarily mediates the entry of Na+ into and the exit of H+ from the proximal tubules, directly and indirectly being responsible for reabsorbing ~50% of filtered Na+ in the proximal tubules of the kidney. On the basolateral membrane side, Na+/K+-ATPase serves as a powerful engine driving Na+ out of, while pumping K+ into the proximal tubules against their concentration gradients. While the roles of NHE3 and Na+/K+-ATPase in proximal tubular Na+ transport under in vitro conditions are well recognized, their respective contributions to the basal blood pressure regulation and angiotensin II (ANG II)-induced hypertension remain poorly understood. Recently, we have been fortunate to be able to use genetically modified mouse models with global, kidney- or proximal tubule-specific deletion of NHE3 to directly determine the cause and effect relationship between NHE3, basal blood pressure homeostasis, and ANG II-induced hypertension at the whole body, kidney and/or proximal tubule levels. The purpose of this article is to review the genetic and genomic evidence for an important role of NHE3 with a focus in the regulation of basal blood pressure and ANG II-induced hypertension, as we learned from studies using global, kidney- or proximal tubule-specific NHE3 knockout mice. We hypothesize that NHE3 in the proximal tubules is necessary for maintaining basal blood pressure homeostasis and the development of ANG II-induced hypertension.


Asunto(s)
Angiotensina II/farmacología , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/genética , Hipertensión/inducido químicamente , Hipertensión/genética , Intercambiador 3 de Sodio-Hidrógeno/genética , Animales , Humanos , Túbulos Renales Proximales/efectos de los fármacos
6.
Clin Sci (Lond) ; 132(13): 1383-1401, 2018 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-29986878

RESUMEN

The renin-angiotensin system (RAS) is widely recognized as one of the most important vasoactive hormonal systems in the physiological regulation of blood pressure and the development of hypertension. This recognition is derived from, and supported by, extensive molecular, cellular, genetic, and pharmacological studies on the circulating (tissue-to-tissue), paracrine (cell-to-cell), and intracrine (intracellular, mitochondrial, nuclear) RAS during last several decades. Now, it is widely accepted that circulating and local RAS may act independently or interactively, to regulate sympathetic activity, systemic and renal hemodynamics, body salt and fluid balance, and blood pressure homeostasis. However, there remains continuous debate with respect to the specific sources of intratubular and intracellular RAS in the kidney and other tissues, the relative contributions of the circulating RAS to intratubular and intracellular RAS, and the roles of intratubular compared with intracellular RAS to the normal control of blood pressure or the development of angiotensin II (ANG II)-dependent hypertension. Based on a lecture given at the recent XI International Symposium on Vasoactive Peptides held in Horizonte, Brazil, this article reviews recent studies using mouse models with global, kidney- or proximal tubule-specific overexpression (knockin) or deletion (knockout) of components of the RAS or its receptors. Although much knowledge has been gained from cell- and tissue-specific transgenic or knockout models, a unifying and integrative approach is now required to better understand how the circulating and local intratubular/intracellular RAS act independently, or with other vasoactive systems, to regulate blood pressure, cardiovascular and kidney function.


Asunto(s)
Presión Sanguínea/fisiología , Riñón/metabolismo , Sistema Renina-Angiotensina/fisiología , Angiotensina II/fisiología , Angiotensinógeno/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Riñón/fisiología , Túbulos Renales Proximales/metabolismo , Hígado/metabolismo , Ratones , Renina/fisiología
7.
Am J Physiol Renal Physiol ; 313(2): F440-F449, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28468964

RESUMEN

ANG II has many biological effects in renal physiology, particularly in Ca2+ handling in the regulation of fluid and solute reabsorption. It involves the systemic endocrine renin-angiotensin system (RAS), but tissue and intracrine ANG II are also known. We have shown that ANG II induces heterodimerization of its AT1 and AT2 receptors (AT1R and AT2R) to stimulate sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) activity. Thus, we investigated whether ANG II-AT1R/AT2R complex is formed and internalized, and also examined the intracellular localization of this complex to determine how its effect might be exerted on renal intracrine RAS. Living cell imaging of LLC-PK1 cells, quantification of extracellular ANG II, and use of the receptor antagonists, losartan and PD123319, showed that ANG II is internalized with AT1R/AT2R heterodimers as a complex in a microtubule-dependent and clathrin-independent manner, since colchicine-but not Pitstop2-blocked this process. This result was confirmed by an increase of ß-arrestin phosphorylation after ANG II treatment, clathrin-mediated endocytosis being dependent on dephosphorylation of ß-arrestin. Internalized ANG II colocalized with an endoplasmic reticulum (ER) marker and increased levels of AT1R, AT2R, and PKCα in ER-enriched membrane fractions. This novel evidence suggests the internalization of an ANG II-AT1/AT2 complex to target ER, where it might trigger intracellular Ca2+ responses.


Asunto(s)
Angiotensina II/metabolismo , Membrana Celular/metabolismo , Endocitosis , Retículo Endoplásmico/metabolismo , Riñón/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Receptor de Angiotensina Tipo 2/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Bloqueadores del Receptor Tipo 2 de Angiotensina II/farmacología , Animales , Calcio/metabolismo , Membrana Celular/efectos de los fármacos , Endocitosis/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Riñón/efectos de los fármacos , Células LLC-PK1 , Microtúbulos/metabolismo , Complejos Multiproteicos , Fosforilación , Proteína Quinasa C-alfa/metabolismo , Transporte de Proteínas , Receptor de Angiotensina Tipo 1/efectos de los fármacos , Receptor de Angiotensina Tipo 2/efectos de los fármacos , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Porcinos , beta-Arrestinas/metabolismo
8.
Pharmacol Res ; 125(Pt A): 21-38, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28619367

RESUMEN

The renin-angiotensin system (RAS) is undisputedly one of the most prominent endocrine (tissue-to-tissue), paracrine (cell-to-cell) and intracrine (intracellular/nuclear) vasoactive systems in the physiological regulation of neural, cardiovascular, blood pressure, and kidney function. The importance of the RAS in the development and pathogenesis of cardiovascular, hypertensive and kidney diseases has now been firmly established in clinical trials and practice using renin inhibitors, angiotensin-converting enzyme (ACE) inhibitors, type 1 (AT1) angiotensin II (ANG II) receptor blockers (ARBs), or aldosterone receptor antagonists as major therapeutic drugs. The major mechanisms of actions for these RAS inhibitors or receptor blockers are mediated primarily by blocking the detrimental effects of the classic angiotensinogen/renin/ACE/ANG II/AT1/aldosterone axis. However, the RAS has expanded from this classic axis to include several other complex biochemical and physiological axes, which are derived from the metabolism of this classic axis. Currently, at least five axes of the RAS have been described, with each having its key substrate, enzyme, effector peptide, receptor, and/or downstream signaling pathways. These include the classic angiotensinogen/renin/ACE/ANG II/AT1 receptor, the ANG II/APA/ANG III/AT2/NO/cGMP, the ANG I/ANG II/ACE2/ANG (1-7)/Mas receptor, the prorenin/renin/prorenin receptor (PRR or Atp6ap2)/MAP kinases ERK1/2/V-ATPase, and the ANG III/APN/ANG IV/IRAP/AT4 receptor axes. Since the roles and therapeutic implications of the classic angiotensinogen/renin/ACE/ANG II/AT1 receptor axis have been extensively reviewed, this article will focus primarily on reviewing the roles and therapeutic implications of the vasoprotective axes of the RAS in cardiovascular, hypertensive and kidney diseases.


Asunto(s)
Enfermedades Cardiovasculares/fisiopatología , Hipertensión/fisiopatología , Enfermedades Renales/fisiopatología , Sistema Renina-Angiotensina/fisiología , Animales , Humanos , Transducción de Señal/fisiología
9.
Am J Physiol Renal Physiol ; 310(10): F1103-12, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26864937

RESUMEN

Long-term angiotensin II (ANG II) infusion significantly increases ANG II levels in the kidney through two major mechanisms: AT1 receptor-mediated augmentation of angiotensinogen (AGT) expression and uptake of circulating ANG II by the proximal tubules. However, it is not known whether intracellular ANG II stimulates AGT expression in the proximal tubule. In the present study, we overexpressed an intracellular cyan fluorescent ANG II fusion protein (Ad-sglt2-ECFP/ANG II) selectively in the proximal tubule of rats and mice using the sodium and glucose cotransporter 2 (sglt2) promoter. AGT mRNA and protein expression in the renal cortex and 24-h urinary AGT excretion were determined 4 wk following overexpression of ECFP/ANG II in the proximal tubule. Systolic blood pressure was significantly increased with a small antinatriuretic effect in rats and mice with proximal tubule-selective expression of ECFP/ANG II (P < 0.01). AGT mRNA and protein expression in the cortex were increased by >1.5-fold and 61 ± 16% (P < 0.05), whereas urinary AGT excretion was increased from 48.7 ± 5.7 (n = 13) to 102 ± 13.5 (n = 13) ng/24 h (P < 0.05). However, plasma AGT, renin activity, and ANG II levels remained unaltered by ECFP/ANG II. The increased AGT mRNA and protein expressions in the cortex by ECFP/ANG II were blocked in AT1a-knockout (KO) mice. Studies in cultured mouse proximal tubule cells demonstrated involvement of AT1a receptor/MAP kinases/NF-кB signaling pathways. These results indicate that intracellular ANG II stimulates AGT expression in the proximal tubules, leading to increased AGT formation and secretion into the tubular fluid, which contributes to ANG II-dependent hypertension.


Asunto(s)
Angiotensina II/metabolismo , Angiotensinógeno/metabolismo , Túbulos Renales Proximales/metabolismo , Sistema de Señalización de MAP Quinasas , Receptor de Angiotensina Tipo 1/metabolismo , Animales , Presión Sanguínea , Hipertensión/metabolismo , Masculino , FN-kappa B/metabolismo , Ratas Sprague-Dawley , Renina/sangre , Sistema Renina-Angiotensina , Sodio/orina
10.
Curr Hypertens Rep ; 18(8): 63, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27372447

RESUMEN

It is well recognized that the renin-angiotensin system (RAS) exists not only as circulating, paracrine (cell to cell), but also intracrine (intracellular) system. In the kidney, however, it is difficult to dissect the respective contributions of circulating RAS versus intrarenal RAS to the physiological regulation of proximal tubular Na(+) reabsorption and hypertension. Here, we review recent studies to provide an update in this research field with a focus on the proximal tubular RAS in angiotensin II (ANG II)-induced hypertension. Careful analysis of available evidence supports the hypothesis that both local synthesis or formation and AT1 (AT1a) receptor- and/or megalin-mediated uptake of angiotensinogen (AGT), ANG I and ANG II contribute to high levels of ANG II in the proximal tubules of the kidney. Under physiological conditions, nearly all major components of the RAS including AGT, prorenin, renin, ANG I, and ANG II would be filtered by the glomerulus and taken up by the proximal tubules. In ANG II-dependent hypertension, the expression of AGT, prorenin, and (pro)renin receptors, and angiotensin-converting enzyme (ACE) is upregulated rather than downregulated in the kidney. Furthermore, hypertension damages the glomerular filtration barrier, which augments the filtration of circulating AGT, prorenin, renin, ANG I, and ANG II and their uptake in the proximal tubules. Together, increased local ANG II formation and augmented uptake of circulating ANG II in the proximal tubules, via activation of AT1 (AT1a) receptors and Na(+)/H(+) exchanger 3, may provide a powerful feedforward mechanism for promoting Na(+) retention and the development of ANG II-induced hypertension.


Asunto(s)
Angiotensina II/fisiología , Hipertensión/fisiopatología , Túbulos Renales Proximales/fisiopatología , Sistema Renina-Angiotensina/fisiología , Angiotensinógeno/fisiología , Animales , Femenino , Humanos , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/fisiología , Masculino , Receptor de Angiotensina Tipo 1/metabolismo , Receptores de Superficie Celular/fisiología , Sodio/sangre , Sodio/metabolismo , Intercambiadores de Sodio-Hidrógeno/fisiología , Regulación hacia Arriba/fisiología , ATPasas de Translocación de Protón Vacuolares/fisiología , Receptor de Prorenina
11.
Physiol Genomics ; 47(10): 479-87, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26242933

RESUMEN

The renal mechanisms responsible for angiotensin II (ANG II)-induced hypertension remain incompletely understood. The present study tested the hypothesis that the Na(+)/H(+) exchanger 3 (NHE3) is required for ANG II-induced hypertension in mice. Five groups of wild-type (Nhe3(+/+)) and Nhe3(-/-) mice were treated with vehicle or high pressor doses of ANG II (1.5 mg/kg/day ip, via minipump for 2 wk, or 10 pmol/min iv for 30 min). Under basal conditions, Nhe3(-/-) mice had significantly lower systolic blood pressure (SBP) and mean intra-arterial pressure (MAP) (P < 0.01), 24 h urine (P < 0.05), urinary Na(+) (P < 0.01) and urinary K(+) excretion (P < 0.01). In response to ANG II, SBP and MAP markedly increased in Nhe3(+/+) mice in a time-dependent manner, as expected (P < 0.01). However, these acute and chronic pressor responses to ANG II were significantly attenuated in Nhe3(-/-) mice (P < 0.01). Losartan blocked ANG II-induced hypertension in Nhe3(+/+) mice but induced marked mortality in Nhe3(-/-) mice. The attenuated pressor responses to ANG II in Nhe3(-/-) mice were associated with marked compensatory humoral and renal responses to genetic loss of intestinal and renal NHE3. These include elevated basal plasma ANG II and aldosterone and kidney ANG II levels, salt wasting from the intestines, increased renal AQP1, Na(+)/HCO3 (-), and Na(+)/K(+)-ATPase expression, and increased PKCα, mitogen-activated protein kinases ERK1/2, and glycogen synthase kinase 3αß signaling proteins in the proximal tubules (P < 0.01). We concluded that NHE3 in proximal tubules of the kidney, along with NHE3 in intestines, is required for maintaining basal blood pressure as well as the full development of ANG II-induced hypertension.


Asunto(s)
Hipertensión/inducido químicamente , Hipertensión/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Aldosterona/sangre , Anestesia , Angiotensina II/sangre , Animales , Presión Sanguínea , Electrólitos/orina , Hipertensión/sangre , Hipertensión/fisiopatología , Intestinos/patología , Riñón/patología , Túbulos Renales/metabolismo , Túbulos Renales/fisiopatología , Masculino , Ratones , Fenotipo , Fosforilación , Potasio/orina , Transducción de Señal , Sodio/orina , Sístole
12.
Am J Physiol Renal Physiol ; 307(2): F222-33, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24740791

RESUMEN

The present study tested the hypothesis that the multiligand endocytic receptor megalin is partially involved in the uptake of ANG II and downstream signaling responses in mouse proximal tubule cells (mPCT) by interacting with AT1a receptors. mPCT cells of wild-type (WT) and AT1a receptor-deficient (AT1a-KO) mice were treated with vehicle, the AT1 receptor blocker losartan (10 µM), or a selective megalin small interfering (si) RNA for 48 h. The uptake of fluorescein (FITC)-labeled ANG II (10 nM, 37°C) and downstream signaling responses were analyzed by fluorescence imaging and Western blotting. AT1a receptors and megalin were abundantly expressed in mPCT cells, whereas AT1a receptors were absent in AT1a-KO mPCT cells (P < 0.01). In WT mPCT cells, FITC-ANG II uptake was visualized at 30 min in the cytoplasm and in the nuclei 1 h after exposure. Losartan alone completely blocked the uptake of FITC-ANG II, whereas megalin siRNA inhibited only 30% of the response (P < 0.01). The remaining FITC-ANG II uptake in the presence of megalin siRNA was completely abolished by losartan. ANG II induced threefold increases in phosphorylated MAP kinases ERK1/2 and a onefold increase in phosphorylated sodium and hydrogen exchanger 3 (NHE3) proteins, which were also blocked by losartan and megalin-siRNA. By contrast, losartan and megalin siRNA had no effects on these signaling proteins in AT1a-KO mPCT cells. We conclude that the uptake of ANG II and downstream MAP kinases ERK1/2 and NHE3 signaling responses in mPCT cells are mediated primarily by AT1a receptors. However, megalin may also play a partial role in these responses to ANG II.


Asunto(s)
Angiotensina II/metabolismo , Endocitosis , Túbulos Renales Proximales/metabolismo , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Angiotensina II/análogos & derivados , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Células Cultivadas , Endocitosis/efectos de los fármacos , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Ligandos , Losartán/farmacología , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Interferencia de ARN , Receptor de Angiotensina Tipo 1/deficiencia , Receptor de Angiotensina Tipo 1/efectos de los fármacos , Receptor de Angiotensina Tipo 1/genética , Transducción de Señal , Intercambiador 3 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/metabolismo , Transfección
13.
Am J Physiol Renal Physiol ; 307(8): F949-61, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25164083

RESUMEN

Caveolin 1 (CAV-1) functions not only as a constitutive scaffolding protein of caveolae but also as a vesicular transporter and signaling regulator. In the present study, we tested the hypothesis that CAV-1 knockout (CAV-1 KO) inhibits ANG II type 1 [AT1 (AT1a)] receptor-mediated uptake of ANG II in the proximal tubule and attenuates blood pressure responses in ANG II-induced hypertension. To determine the role of CAV-1 in mediating the uptake of FITC-labeled ANG II, wild-type (WT) mouse proximal convoluted tubule cells were transfected with CAV-1 small interfering (si)RNA for 48 h before AT1 receptor-mediated uptake of FITC-labeled ANG II was studied. CAV-1 siRNA knocked down CAV-1 expression by >90% (P < 0.01) and inhibited FITC-labeled ANG II uptake by >50% (P < 0.01). Moreover, CAV-1 siRNA attenuated ANG II-induced activation of MAPK ERK1/2 and Na(+)/H(+) exchanger 3 expression, respectively (P < 0.01). To determine whether CAV-1 regulates ANG II uptake in the proximal tubule, Alexa 488-labeled ANG II was infused into anesthetized WT and CAV-1 KO mice for 60 min (20 ng/min iv). Imaging analysis revealed that Alexa 488-labeled ANG II uptake was decreased by >50% in CAV-1 KO mice (P < 0.01). Furthermore, Val(5)-ANG II was infused into WT and CAV-1 KO mice for 2 wk (1.5 mg·kg(-1)·day(-1) ip). Basal systolic pressure was higher, whereas blood pressure and renal excretory and signaling responses to ANG II were attenuated, in CAV-1 KO mice (P < 0.01). We concluded that CAV-1 plays an important role in AT1 receptor-mediated uptake of ANG II in the proximal tubule and modulates blood pressure and renal responses to ANG II.


Asunto(s)
Angiotensina II/metabolismo , Caveolina 1/fisiología , Túbulos Renales Proximales/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Caveolina 1/genética , Células Cultivadas , Túbulos Renales Proximales/citología , Masculino , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , ARN Interferente Pequeño/metabolismo , Intercambiador 3 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/biosíntesis
14.
Am J Physiol Regul Integr Comp Physiol ; 304(8): R588-98, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23427083

RESUMEN

The role of intracellular ANG II in proximal tubules of the kidney remains poorly understood. We tested the hypothesis that proximal tubule-dominant transfer of AT(1a) receptors in the cortex mediates intracellular ANG II-induced blood pressure responses in AT(1a) receptor-deficient (Agtr1a-/-) mice. A GFP-tagged AT(1a) receptor, AT(1a)R/GFP, and an enhanced cyan fluorescent intracellular ANG II fusion protein, ECFP/ANG II, were expressed in proximal tubules of Agtr1a-/- mouse kidneys via the adenoviral transfer using a sodium and glucose cotransporter 2 promoter. Transfer of AT(1a)R/GFP alone or with ECFP/ANG II induced proximal tubule-dominant expression of AT(1a)R/GFP and/or ECFP/ANG II with a peak response at 2 wk. No significant AT(1a)R/GFP and/or ECFP/ANG II expression was observed in the glomeruli, medulla, or extrarenal tissues. Transfer of AT(1a)R/GFP alone, but not ECFP/ANG II, increased systolic blood pressure by 12 ± 2 mmHg by day 14 (n = 9, P < 0.01). However, cotransfer of AT(1a)R/GFP with ECFP/ANG II increased blood pressure by 18 ± 2 mmHg (n = 12, P < 0.01). Twenty-four hour urinary sodium excretion was decreased by day 7 with proximal tubule-dominant transfer of AT(1a)R/GFP alone (P < 0.01) or with AT(1a)R/GFP and ECFP/ANG II cotransfer (P < 0.01). These responses were associated with twofold increases in phosphorylated ERK1/2, lysate, and membrane NHE-3 proteins in freshly isolated proximal tubules (P < 0.01). By contrast, transfer of control CMV-GFP (a recombinant human adenovirus type 5 expresses enhanced green fluorescent protein under the control of a cytomegalovirus (CMV) promoter), ECFP/ANG II, or a scrambled control ECFP/ANG IIc alone in proximal tubules had no effect on all indices. These results suggest that AT(1a) receptors and intracellular ANG II in proximal tubules of the kidney play an important physiological role in blood pressure regulation.


Asunto(s)
Angiotensina II/farmacología , Presión Sanguínea/fisiología , Túbulos Renales Proximales/metabolismo , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Vasoconstrictores/farmacología , Angiotensina II/biosíntesis , Animales , Presión Sanguínea/efectos de los fármacos , Western Blotting , Dependovirus , Ingestión de Líquidos/fisiología , Electrólitos/orina , Ensayo de Inmunoadsorción Enzimática , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Túbulos Renales Proximales/efectos de los fármacos , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/biosíntesis , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Angiotensina Tipo 1/fisiología , Transportador 2 de Sodio-Glucosa/metabolismo , Intercambiador 3 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/biosíntesis , Intercambiadores de Sodio-Hidrógeno/genética
15.
Curr Hypertens Rep ; 15(5): 522-30, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23996678

RESUMEN

Type 2 diabetes is well recognized as a noninsulin-dependent diabetic disease. Clinical evidence indicates that the level of circulating insulin may be normal, subnormal, and even elevated in type 2 diabetic patients. Unlike type 1 diabetes, the key problem for type 2 diabetes is not due to the absolute deficiency of insulin secretion, but because the body is no longer sensitive to insulin. Thus, insulin resistance is increased and the sensitivity to insulin is reset, so increasing levels of insulin are required to maintain body glucose and metabolic homeostasis. How insulin resistance is increased and what factors contribute to its development in type 2 diabetes remain incompletely understood. Overemphasis of insulin deficiency alone may be too simplistic for us to understand how type 2 diabetes is developed and should be treated, since glucose metabolism and homeostasis are tightly controlled by both insulin and glucagon. Insulin acts as a YIN factor to lower blood glucose level by increasing cellular glucose uptake, whereas glucagon acts as a YANG factor to counter the action of insulin by increasing glucose production. Furthermore, other humoral factors other than insulin and glucagon may also directly or indirectly contribute to increased insulin resistance and the development of hyperglycemia. The purpose of this article is to briefly review recently published animal and human studies in this field, and provide new insights and perspectives on recent debates as to whether hyperglucagonemia and/or glucagon receptors should be targeted to treat insulin resistance and target organ injury in type 2 diabetes.


Asunto(s)
Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Glucagón/metabolismo , Hiperglucemia/tratamiento farmacológico , Hipertensión/tratamiento farmacológico , Animales , Glucemia/metabolismo , Humanos , Insulina/metabolismo
16.
Can J Cardiol ; 39(12): 1795-1807, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37394059

RESUMEN

The renin-angiotensin system (RAS) is an essential hormonal system involved in water and sodium reabsorption, renal blood flow regulation, and arterial constriction. Systemic stimulation of the RAS with infusion of the main peptide angiotensin II (Ang II) in animals as well as pathological elevation of renin (ie, renovascular hypertension) to increase circulatory Ang II in humans ultimately lead to hypertension and end organ damage. In addition to hypertension, accumulating evidence supports that the Ang II type 1 receptor exerts a critical role in cardiovascular and kidney diseases independent of blood pressure elevation. In the past 2 decades, the identification of an increased number of peptides and receptors has facilitated the concept that the RAS has detrimental and beneficial effects on the cardiovascular system depending on which RAS components are activated. For example, angiotensin 1-7 and Ang II type 2 receptors act as a counter-regulatory system against the classical RAS by mediating vasodilation. Although the RAS as an endocrine system for regulation of blood pressure is well established, there remain many unanswered questions and controversial findings regarding blood pressure regulation and pathophysiological regulation of cardiovascular diseases at the tissue level. This review article includes the latest knowledge gleaned from cell type-selective gene deleted mice regarding cell type-specific roles of Ang II receptors and their significance in health and diseases are discussed. In particular, we focus on the roles of these receptors expressed in vascular, cardiac, and kidney epithelial cells.


Asunto(s)
Hipertensión , Enfermedades Renales , Ratones , Humanos , Animales , Sistema Renina-Angiotensina/fisiología , Hipertensión/genética , Renina , Angiotensina II/metabolismo , Presión Sanguínea
17.
Cells ; 12(11)2023 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-37296603

RESUMEN

Most patients with end-stage renal disease (ESRD) and advanced chronic kidney disease (CKD) choose hemodialysis as their treatment of choice. Thus, upper-extremity veins provide a functioning arteriovenous access to reduce dependence on central venous catheters. However, it is unknown whether CKD reprograms the transcriptome of veins and primes them for arteriovenous fistula (AVF) failure. To examine this, we performed transcriptomic analyses of bulk RNA sequencing data of veins isolated from 48 CKD patients and 20 non-CKD controls and made the following findings: (1) CKD converts veins into immune organs by upregulating 13 cytokine and chemokine genes, and over 50 canonical and noncanonical secretome genes; (2) CKD increases innate immune responses by upregulating 12 innate immune response genes and 18 cell membrane protein genes for increased intercellular communication, such as CX3CR1 chemokine signaling; (3) CKD upregulates five endoplasmic reticulum protein-coding genes and three mitochondrial genes, impairing mitochondrial bioenergetics and inducing immunometabolic reprogramming; (4) CKD reprograms fibrogenic processes in veins by upregulating 20 fibroblast genes and 6 fibrogenic factors, priming the vein for AVF failure; (5) CKD reprograms numerous cell death and survival programs; (6) CKD reprograms protein kinase signal transduction pathways and upregulates SRPK3 and CHKB; and (7) CKD reprograms vein transcriptomes and upregulates MYCN, AP1, and 11 other transcription factors for embryonic organ development, positive regulation of developmental growth, and muscle structure development in veins. These results provide novel insights on the roles of veins as immune endocrine organs and the effect of CKD in upregulating secretomes and driving immune and vascular cell differentiation.


Asunto(s)
Derivación Arteriovenosa Quirúrgica , Insuficiencia Renal Crónica , Humanos , Proteína Proto-Oncogénica N-Myc/metabolismo , Derivación Arteriovenosa Quirúrgica/métodos , Venas , Insuficiencia Renal Crónica/metabolismo , Transducción de Señal
18.
Am J Physiol Renal Physiol ; 303(5): F746-56, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22739536

RESUMEN

It is well recognized that ANG II interacts with arginine vasopressin (AVP) to regulate water reabsorption and urine concentration in the kidney. The present study used ANG II type 1a (AT(1a)) receptor-deficient (Agtr1a(-/-)) mice to test the hypothesis that AT(1a) receptor signaling is required for basal and water deprivation-induced urine concentration in the renal medulla. Eight groups of wild-type (WT) and Agtr1a(-/-) mice were treated with or without 24-h water deprivation and 1-desamino-8-d-AVP (DDAVP; 100 ng/h ip) for 2 wk or with losartan (10 mg/kg ip) during water deprivation. Under basal conditions, Agtr1a(-/-) mice had lower systolic blood pressure (P < 0.01), greater than threefold higher 24-h urine excretion (WT mice: 1.3 ± 0.1 ml vs. Agtr1a(-/-) mice: 5.9 ± 0.7 ml, P < 0.01), and markedly decreased urine osmolality (WT mice: 1,834 ± 86 mosM/kg vs. Agtr1a(-/-) mice: 843 ± 170 mosM/kg, P < 0.01), without significant changes in 24-h urinary Na(+) excretion. These responses in Agtr1a(-/-) mice were associated with lower basal plasma AVP (WT mice: 105 ± 8 pg/ml vs. Agtr1a(-/-) mice: 67 ± 6 pg/ml, P < 0.01) and decreases in total lysate and membrane aquaporin-2 (AQP2; 48.6 ± 7% of WT mice, P < 0.001) and adenylyl cyclase isoform III (55.6 ± 8% of WT mice, P < 0.01) proteins. Although 24-h water deprivation increased plasma AVP to the same levels in both strains, 24-h urine excretion was still higher, whereas urine osmolality remained lower, in Agtr1a(-/-) mice (P < 0.01). Water deprivation increased total lysate AQP2 proteins in the inner medulla but had no effect on adenylyl cyclase III, phosphorylated MAPK ERK1/2, and membrane AQP2 proteins in Agtr1a(-/-) mice. Furthermore, infusion of DDAVP for 2 wk was unable to correct the urine-concentrating defects in Agtr1a(-/-) mice. These results demonstrate that AT(1a) receptor-mediated ANG II signaling is required to maintain tonic AVP release and regulate V(2) receptor-mediated responses to water deprivation in the inner medulla.


Asunto(s)
Receptor de Angiotensina Tipo 1/deficiencia , Receptor de Angiotensina Tipo 1/fisiología , Adenilil Ciclasas/metabolismo , Animales , Acuaporina 2/metabolismo , Arginina Vasopresina/sangre , Presión Sanguínea , Desamino Arginina Vasopresina/farmacología , Losartán/farmacología , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Concentración Osmolar , Privación de Agua
19.
Am J Physiol Regul Integr Comp Physiol ; 302(5): R494-509, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22170616

RESUMEN

ANG II is the most potent and important member of the classical renin-angiotensin system (RAS). ANG II, once considered to be an endocrine hormone, is now increasingly recognized to also play novel and important paracrine (cell-to-cell) and intracrine (intracellular) roles in cardiovascular and renal physiology and blood pressure regulation. Although an intracrine role of ANG II remains an issue of continuous debates and requires further confirmation, a great deal of research has recently been devoted to uncover the novel actions and elucidate underlying signaling mechanisms of the so-called intracellular ANG II in cardiovascular, neural, and renal systems. The purpose of this article is to provide a comprehensive review of the intracellular actions of ANG II, either administered directly into the cells or expressed as an intracellularly functional fusion protein, and its effects throughout a variety of target tissues susceptible to the impacts of an overactive ANG II, with a particular focus on the proximal tubules of the kidney. While continuously reaffirming the roles of extracellular or circulating ANG II in the proximal tubules, our review will focus on recent evidence obtained for the novel biological roles of intracellular ANG II in cultured proximal tubule cells in vitro and the potential physiological roles of intracellular ANG II in the regulation of proximal tubular reabsorption and blood pressure in rats and mice. It is our hope that the new knowledge on the roles of intracellular ANG II in proximal tubules will serve as a catalyst to stimulate further studies and debates in the field and to help us better understand how extracellular and intracellular ANG II acts independently or interacts with each other, to regulate proximal tubular transport and blood pressure in both physiological and diseased states.


Asunto(s)
Angiotensina II/fisiología , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/fisiología , Sistema Renina-Angiotensina/fisiología , Animales , Presión Sanguínea/fisiología , Núcleo Celular/fisiología , Citoplasma/fisiología , Ratones , Modelos Animales , Ratas , Receptor de Angiotensina Tipo 1/fisiología
20.
Am J Physiol Renal Physiol ; 300(5): F1076-88, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21307128

RESUMEN

The present study tested the hypothesis that intrarenal adenoviral transfer of an intracellular cyan fluorescent fusion of angiotensin II (ECFP/ANG II) selectively in proximal tubules of the kidney increases blood pressure by activating AT(1) (AT(1a)) receptors. Intrarenal transfer of ECFP/ANG II was induced in the superficial cortex of rat and mouse kidneys, and the sodium and glucose cotransporter 2 (sglt2) promoter was used to drive ECFP/ANG II expression selectively in proximal tubules. Intrarenal transfer of ECFP/ANG II induced a time-dependent, proximal tubule-selective expression of ECFP/ANG II in the cortex, which peaked at 2 wk and was sustained for 4 wk. ECFP/ANG II expression was low in the glomeruli and the entire medulla and was absent in the contralateral kidney or extrarenal tissues. At its peak of expression in proximal tubules at day 14, ANG II was increased by twofold in the kidney (P < 0.01) and more than threefold in proximal tubules (P < 0.01), but remained unchanged in plasma or urine. Systolic blood pressure was increased in ECFP/ANG II-transferred rats by 28 ± 6 mmHg (P < 0.01), whereas fractional sodium excretion was decreased by 20% (P < 0.01) and fractional lithium excretion was reduced by 24% (P < 0.01). These effects were blocked by losartan and prevented in AT(1a) knockout mice. Transfer of a scrambled ECFP/ANG IIc had no effects on blood pressure, kidney, and proximal tubule ANG II, or sodium excretion. These results provide evidence that proximal tubule-selective transfer of an intracellular ANG II fusion protein increases blood pressure by activating AT(1a) receptors and increasing sodium reabsorption in proximal tubules.


Asunto(s)
Angiotensina II/biosíntesis , Presión Sanguínea , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/biosíntesis , Hipertensión/metabolismo , Túbulos Renales Proximales/metabolismo , Adenoviridae/genética , Angiotensina II/sangre , Angiotensina II/genética , Angiotensina II/orina , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/genética , Células Cultivadas , Modelos Animales de Enfermedad , Vectores Genéticos , Proteínas Fluorescentes Verdes/sangre , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/orina , Hipertensión/genética , Hipertensión/fisiopatología , Hipertensión/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Natriuresis , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1/deficiencia , Receptor de Angiotensina Tipo 1/efectos de los fármacos , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Factores de Tiempo , Transducción Genética , Transfección , Regulación hacia Arriba , Micción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA