Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Pharm Res ; 41(5): 849-861, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38485855

RESUMEN

PURPOSE: Olmesartan medoxomil (olmesartan-MX), an ester-type prodrug of the angiotensin II receptor blocker (ARB) olmesartan, is predominantly anionic at intestinal pH. Human organic anion transporting polypeptide 2B1 (OATP2B1) is expressed in the small intestine and is involved in the absorption of various acidic drugs. This study was designed to test the hypothesis that OATP2B1-mediated uptake contributes to the enhanced intestinal absorption of olmesartan-MX, even though olmesartan itself is not a substrate of OATP2B1. METHODS: Tetracycline-inducible human OATP2B1- and rat Oatp2b1-overexpressing HEK 293 cell lines (hOATP2B1/T-REx-293 and rOatp2b1/T-REx-293, respectively) were established to characterize OATP2B1-mediated uptake. Rat jejunal permeability was measured using Ussing chambers. ARBs were quantified by liquid chromatography-tandem mass spectrometry. RESULTS: Significant olmesartan-MX uptake was observed in hOATP2B1/T-REx-293 and rOatp2b1/T-REx-293 cells, whereas olmesartan uptake was undetectable or much lower than olmesartan-MX uptake, respectively. Furthermore, olmesartan-MX exhibited several-fold higher uptake in Caco-2 cells and greater permeability in rat jejunum compared to olmesartan. Olmesartan-MX uptake in hOATP2B1/T-REx-293 cells and in Caco-2 cells was significantly decreased by OATP2B1 substrates/inhibitors such as 1 mM estrone-3-sulfate, 100 µM rifamycin SV, and 100 µM fluvastatin. Rat Oatp2b1-mediated uptake and rat jejunal permeability of olmesartan-MX were significantly decreased by 50 µM naringin, an OATP2B1 inhibitor. Oral administration of olmesartan-MX with 50 µM naringin to rats significantly reduced the area under the plasma concentration-time curve of olmesartan to 76.9%. CONCLUSION: Olmesartan-MX is a substrate for OATP2B1, and the naringin-sensitive transport system contributes to the improved intestinal absorption of olmesartan-MX compared with its parent drug, olmesartan.


Asunto(s)
Imidazoles , Absorción Intestinal , Olmesartán Medoxomilo , Transportadores de Anión Orgánico , Profármacos , Tetrazoles , Animales , Humanos , Absorción Intestinal/efectos de los fármacos , Olmesartán Medoxomilo/metabolismo , Profármacos/farmacocinética , Profármacos/metabolismo , Células HEK293 , Tetrazoles/farmacocinética , Tetrazoles/metabolismo , Transportadores de Anión Orgánico/metabolismo , Transportadores de Anión Orgánico/antagonistas & inhibidores , Masculino , Imidazoles/farmacocinética , Imidazoles/metabolismo , Ratas , Ratas Sprague-Dawley , Yeyuno/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacocinética , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Permeabilidad/efectos de los fármacos , Células CACO-2
2.
Horm Metab Res ; 52(6): 379-385, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32168525

RESUMEN

Primary aldosteronism (PA) is the most common form of endocrine hypertension. Agonistic autoantibodies against the angiotensin II type 1 receptor (AT1R-Abs) have been described in transplantation medicine and women with pre-eclampsia and more recently in patients with PA. Any functional role of AT1R-Abs in either of the two main subtypes of PA (aldosterone-producing adenoma or bilateral adrenal hyperplasia) requires clarification. In this review, we discuss the studies performed to date on AT1R-Abs in PA.


Asunto(s)
Autoanticuerpos/fisiología , Hiperaldosteronismo/etiología , Hiperaldosteronismo/inmunología , Receptor de Angiotensina Tipo 1/inmunología , Neoplasias de la Corteza Suprarrenal/complicaciones , Neoplasias de la Corteza Suprarrenal/inmunología , Neoplasias de la Corteza Suprarrenal/metabolismo , Adenoma Corticosuprarrenal/complicaciones , Adenoma Corticosuprarrenal/inmunología , Adenoma Corticosuprarrenal/metabolismo , Aldosterona/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Autoanticuerpos/metabolismo , Femenino , Humanos , Hipertensión/etiología , Hipertensión/metabolismo , Masculino , Preeclampsia/etiología , Preeclampsia/metabolismo , Embarazo
3.
Sci Rep ; 9(1): 16205, 2019 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-31700033

RESUMEN

Angiotensin II type-1 receptor-neprilysin inhibitor (ARNi) is consisted of Angiotensin II type-1 receptor (AT1) antagonist and neprilysin (NEP) inhibitor, which could simultaneously increase the vasodilators of the natriuretic peptides and antagonize vasoconstrictors of Ang II. ARNi has been proved a superior effect and lower risks of death on chronic heart failure (CHF) and hypertension. In this paper, ARNi from Traditional Chinese Medicines (TCM) was discovered based on target combination of AT1 and NEP by virtual screening, biological assay and molecular dynamics (MD) simulations. Two customized strategies of combinatorial virtual screening were implemented to discover AT1 antagonist and NEP inhibitor based on pharmacophore modeling and docking computation respectively. Gyrophoric acid (PubChem CID: 135728) from Parmelia saxatilis was selected as AT1 antagonist and assayed with IC50 of 29.76 µM by calcium influx assay. And 3,5,3'-triiodothyronine (PubChem CID: 861) from Bos taurus domesticus was screened as NEP inhibitor and has a dose dependent inhibitory activity by biochemistry fluorescence assay. Combined with MD simulations, these compounds can generate interaction with the target, key interactive residues of ARG167, TRP84, and VAL108 in AT1, and HIS711 in NEP were also identified respectively. This study designs the combinatorial strategy to discover novel frames of ARNi from TCM, and gyrophoric acid and 3,5,3'-triiodothyronine could provide the clues and revelations of drug design and therapeutic method of CHF and hypertension for TCM clinical applications.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Descubrimiento de Drogas , Medicina Tradicional China , Neprilisina/antagonistas & inhibidores , Receptor de Angiotensina Tipo 1/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Enfermedad Crónica , Evaluación Preclínica de Medicamentos , Insuficiencia Cardíaca/tratamiento farmacológico , Hipertensión/tratamiento farmacológico , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Terapia Molecular Dirigida , Neprilisina/química , Neprilisina/metabolismo , Conformación Proteica , Receptor de Angiotensina Tipo 1/química
4.
Cell ; 176(3): 479-490.e12, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30639100

RESUMEN

The angiotensin II (AngII) type 1 receptor (AT1R) is a critical regulator of cardiovascular and renal function and is an important model for studies of G-protein-coupled receptor (GPCR) signaling. By stabilizing the receptor with a single-domain antibody fragment ("nanobody") discovered using a synthetic yeast-displayed library, we determined the crystal structure of active-state human AT1R bound to an AngII analog with partial agonist activity. The nanobody binds to the receptor's intracellular transducer pocket, stabilizing the large conformational changes characteristic of activated GPCRs. The peptide engages the AT1R through an extensive interface spanning from the receptor core to its extracellular face and N terminus, remodeling the ligand-binding cavity. Remarkably, the mechanism used to propagate conformational changes through the receptor diverges from other GPCRs at several key sites, highlighting the diversity of allosteric mechanisms among GPCRs. Our structure provides insight into how AngII and its analogs stimulate full or biased signaling, respectively.


Asunto(s)
Receptor de Angiotensina Tipo 1/metabolismo , Anticuerpos de Dominio Único/farmacología , Angiotensina II , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Arrestinas/metabolismo , Células HEK293 , Humanos , Fragmentos de Inmunoglobulinas/farmacología , Conformación Proteica , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Anticuerpos de Dominio Único/metabolismo , beta-Arrestinas/metabolismo
5.
Biochem Pharmacol ; 154: 104-117, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29684376

RESUMEN

G protein coupled receptors (GPCRs) produce pleiotropic effects by their capacity to engage numerous signaling pathways once activated. Functional selectivity (also called biased signaling), where specific compounds can bring GPCRs to adopt conformations that enable selective receptor coupling to distinct signaling pathways, continues to be significantly investigated. However, an important but often overlooked aspect of functional selectivity is the capability of ligands such as angiotensin II (AngII) to adopt specific conformations that may preferentially bind to selective GPCRs structures. Understanding both receptor and ligand conformation is of the utmost importance for the design of new drugs targeting GPCRs. In this study, we examined the properties of AngII cyclic analogs to impart biased agonism on the angiotensin type 1 receptor (AT1R). Positions 3 and 5 of AngII were substituted for cysteine and homocysteine residues ([Sar1Hcy3,5]AngII, [Sar1Cys3Hcy5]AngII and [Sar1Cys3,5]AngII) and the resulting analogs were evaluated for their capacity to activate the Gq/11, G12, Gi2, Gi3, Gz, ERK and ß-arrestin (ßarr) signaling pathways via AT1R. Interestingly, [Sar1Hcy3,5]AngII exhibited potency and full efficacy on all pathways tested with the exception of the Gq pathway. Molecular dynamic simulations showed that the energy barrier associated with the insertion of residue Phe8 of AngII within the hydrophobic core of AT1R, associated with Gq/11 activation, is increased with [Sar1Hcy3,5]AngII. These results suggest that constraining the movements of molecular determinants within a given ligand by introducing cyclic structures may lead to the generation of novel ligands providing more efficient biased agonism.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Angiotensina II/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Transducción de Señal/efectos de los fármacos , Angiotensina II/química , Angiotensina II/farmacología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/química , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Receptor de Angiotensina Tipo 1/química , Transducción de Señal/fisiología
6.
Bioorg Med Chem Lett ; 27(16): 3621-3628, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28666735

RESUMEN

While historically 'in vitro' binding data were obtained by analyzing equilibrium experiments, kinetic data are increasingly appreciated to provide information on the time a particular compound remains bound to its target. This information is of biological importance to understand the molecular mechanism of a drug not only to evaluate the time a particular receptor/enzyme is blocked in the case of antagonists/inhibitors but also to investigate its contribution to the efficacy to mediate signaling in the case of agonists. There is accumulating evidence that many drugs binding to either membrane-bound receptors or enzymes are found to display long duration of action which can be ascribed to slow dissociation from their target proteins. In the present review three such examples are discussed which encompass ligands that bind to membrane-bound proteins and from which it appears that the tight binding kinetics is influenced by the cellular/membrane environment of the target protein.


Asunto(s)
Ligandos , Receptores de Superficie Celular/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/química , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Dipeptidil Peptidasa 4/química , Dipeptidil Peptidasa 4/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/química , Inhibidores de la Dipeptidil-Peptidasa IV/metabolismo , Humanos , Cinética , Unión Proteica , Receptor de Angiotensina Tipo 1/química , Receptor de Angiotensina Tipo 1/metabolismo , Receptores de Superficie Celular/agonistas , Receptores de Superficie Celular/antagonistas & inhibidores , Transducción de Señal
7.
Clin Pharmacol Ther ; 101(2): 254-263, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27542885

RESUMEN

Natriuretic peptide (NP) deficiency and sustained renin-angiotensin system activation are associated with impaired oxidative metabolism and predispose to type-2 diabetes. We hypothesized that sacubitril/valsartan (LCZ696), which augments NP through neprilysin inhibition while blocking angiotensin II type-1 (AT1 )-receptors, improves insulin sensitivity, lipid mobilization, and oxidation. After 8 weeks of treatment of obese patients with hypertension, sacubitril/valsartan 400 mg q.d., but not amlodipine 10 mg q.d., was associated with a significant increase from baseline in the insulin sensitivity index (hyperinsulinemic-euglycemic clamp), and tended to be higher in patients treated with sacubitril/valsartan compared to amlodipine. Abdominal adipose tissue interstitial glycerol concentrations increased with sacubitril/valsartan, but decreased with amlodipine. Whole-body lipolysis and substrate oxidation did not change with either treatment. Results confirm that sacubitril/valsartan treatment leads to a metabolic benefit in the study population and supports the relevance of neprilysin inhibition along with AT1 -receptor blockade in the regulation of human glucose and lipid metabolism.


Asunto(s)
Aminobutiratos/farmacología , Antihipertensivos/farmacología , Resistencia a la Insulina/fisiología , Neprilisina/antagonistas & inhibidores , Obesidad/metabolismo , Tetrazoles/farmacología , Tejido Adiposo/efectos de los fármacos , Adulto , Aminobutiratos/uso terapéutico , Amlodipino/farmacología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Compuestos de Bifenilo , Combinación de Medicamentos , Metabolismo Energético/efectos de los fármacos , Femenino , Glicerol/análisis , Humanos , Hipertensión/tratamiento farmacológico , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Persona de Mediana Edad , Péptidos Natriuréticos/genética , Péptidos Natriuréticos/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/fisiología , Tetrazoles/uso terapéutico , Valsartán
8.
Xenobiotica ; 46(11): 986-1000, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26931777

RESUMEN

1. Sacubitril/valsartan (LCZ696) is an angiotensin receptor neprilysin inhibitor (ARNI) providing simultaneous inhibition of neprilysin (neutral endopeptidase 24.11; NEP) and blockade of the angiotensin II type-1 (AT1) receptor. 2. Following oral administration, [(14)C]LCZ696 delivers systemic exposure to valsartan and AHU377 (sacubitril), which is rapidly metabolized to LBQ657 (M1), the biologically active neprilysin inhibitor. Peak sacubitril plasma concentrations were reached within 0.5-1 h. The mean terminal half-lives of sacubitril, LBQ657 and valsartan were ∼1.3, ∼12 and ∼21 h, respectively. 3. Renal excretion was the dominant route of elimination of radioactivity in human. Urine accounted for 51.7-67.8% and feces for 36.9 to 48.3 % of the total radioactivity. The majority of the drug was excreted as the active metabolite LBQ657 in urine and feces, total accounting for ∼85.5% of the total dose. 4. Based upon in vitro studies, the potential for LCZ696 to inhibit or induce cytochrome P450 (CYP) enzymes and cause CYP-mediated drug interactions clinically was found to be low.


Asunto(s)
Aminobutiratos/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Tetrazoles/metabolismo , Valsartán/metabolismo , Adulto , Compuestos de Bifenilo , Combinación de Medicamentos , Humanos , Neprilisina/antagonistas & inhibidores
9.
J Hypertens ; 34(4): 654-65, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26828783

RESUMEN

AIMS: Increasing evidence supports a role for the angiotensin II-AT1-receptor axis in aneurysm development. Here, we studied whether counteracting this axis via stimulation of AT2 receptors is beneficial. Such stimulation occurs naturally during AT1-receptor blockade with losartan, but not during renin inhibition with aliskiren. METHODS AND RESULTS: Aneurysmal homozygous fibulin-4 mice, displaying a four-fold reduced fibulin-4 expression, were treated with placebo, losartan, aliskiren, or the ß-blocker propranolol from day 35 to 100. Their phenotype includes cystic media degeneration, aortic regurgitation, left ventricular dilation, reduced ejection fraction, and fractional shortening. Although losartan and aliskiren reduced hemodynamic stress and increased renin similarly, only losartan increased survival. Propranolol had no effect. No drug rescued elastic fiber fragmentation in established aneurysms, although losartan did reduce aneurysm size. Losartan also increased ejection fraction, decreased LV diameter, and reduced cardiac pSmad2 signaling. None of these effects were seen with aliskiren or propranolol. Longitudinal micro-CT measurements, a novel method in which each mouse serves as its own control, revealed that losartan reduced LV growth more than aneurysm growth, presumably because the heart profits both from the local (cardiac) effects of losartan and its effects on aortic root remodeling. CONCLUSION: Losartan, but not aliskiren or propranolol, improved survival in fibulin-4 mice. This most likely relates to its capacity to improve structure and function of both aorta and heart. The absence of this effect during aliskiren treatment, despite a similar degree of blood pressure reduction and renin-angiotensin system blockade, suggests that it might be because of AT2-receptor stimulation.


Asunto(s)
Aneurisma/fisiopatología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Proteínas de la Matriz Extracelular , Insuficiencia Cardíaca/fisiopatología , Receptor de Angiotensina Tipo 1/metabolismo , Renina/metabolismo , Animales , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Ratones , Ratones Transgénicos , Renina/antagonistas & inhibidores
10.
J Pharm Sci ; 104(9): 3128-35, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25820021

RESUMEN

Mechanisms regulating fetal transfer of olmesartan, an angiotensin-II receptor type 1 antagonist, are important as potential determinants of life-threatening adverse fetal effects. The purpose of this study was to examine the olmesartan transport mechanism through the basal plasma membrane (BM) of human syncytiotrophoblasts forming the placental barrier. Uptake of olmesartan by human placental BM vesicles was potently inhibited by dehydroepiandrosterone sulfate (DHEAS), estrone 3-sulfate, and bromosulfophthalein, which are all typical substrates of organic anion transporter (OAT) 4 localized at the BM of syncytiotrophoblasts, and was increased in the absence of chloride. In tetracycline-inducible OAT4-expressing cells, [(3) H]olmesartan uptake was increased by tetracycline treatment. Olmesartan uptake via OAT4 was concentration dependent with a Km of 20 µM, and was increased in the absence of chloride. [(3) H]Olmesartan efflux via OAT4 was also observed and was trans-stimulated by extracellular chloride and DHEAS. Thus, OAT4 mediates bidirectional transport of olmesartan and appears to regulate fetal transfer of olmesartan at the BM of syncytiotrophoblasts. Efflux transport of olmesartan via OAT4 from syncytiotrophoblasts to the fetal circulation might be facilitated in the presence of an inwardly directed physiological chloride gradient and extracellular DHEAS.


Asunto(s)
Membrana Celular/metabolismo , Imidazoles/metabolismo , Transportadores de Anión Orgánico/metabolismo , Placenta/metabolismo , Tetrazoles/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Membrana Celular/efectos de los fármacos , Cloruros/metabolismo , Sulfato de Deshidroepiandrosterona/farmacología , Estrona/análogos & derivados , Estrona/farmacología , Femenino , Humanos , Placenta/efectos de los fármacos , Embarazo , Sulfobromoftaleína/farmacología , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo
11.
Biomaterials ; 35(29): 8450-66, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24997481

RESUMEN

A multifunctional copolymer-anticancer conjugate chitosan-graft-polyethyleneimine-candesartan (CPC) containing low molecular weight chitosan (CS) backbone and polyethyleneimine (PEI) arms with candesartan (CD) conjugated via an amide bond was fabricated as a targeted co-delivery nanovector of drug and gene for potential cancer therapy. Here, CD was utilized to specifically bind to overexpressed angiotensin II type 1 receptor (AT1R) of tumor cells, strengthen endosomal buffering capacity of CPC and suppress tumor angiogenesis. The self-assembled CPC/pDNA complexes exhibited desirable and homogenous particle size, moderate positive charges, superior stability, and efficient release of drug and gene in vitro. Flow cytometry and confocal laser scanning microscopy analyses confirmed that CD-targeted function and CD-enhanced buffering capacity induced high transfection, specific cellular uptake and efficient intracellular delivery of CPC/pDNA complexes in AT1R-overexpressed PANC-1 cells. In addition, CPC/wt-p53 complexes co-delivering CD and wild type p53 (wt-p53) gene achieved synergistic angiogenesis suppression by more effectively downregulating the expression of vascular endothelial growth factor (VEGF) mRNA and protein via different pathways in vitro, as compared to mono-delivery and mixed-delivery systems. In vivo investigation on nude mice bearing PANC-1 tumor xenografts revealed that CPC/wt-p53 complexes possessed high tumor-targeting capacity and strong anti-tumor activity. Additional analysis of microvessel density (MVD) demonstrated that CPC/wt-p53 complexes significantly inhibited tumor-associated angiogenesis. These findings suggested that CPC could be an ideal tumor-targeting nanovector for simultaneous transfer of drug and gene, and a multifunctional CPC/wt-p53 co-delivery system with tumor-specific targetability, enhanced endosomal buffering capacity and synergistic anti-angiogenesis efficacy might be a new promising strategy for effective tumor therapy.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Bencimidazoles/uso terapéutico , Quitosano/análogos & derivados , ADN/administración & dosificación , Genes p53 , Neoplasias/terapia , Neovascularización Patológica/terapia , Polietileneimina/análogos & derivados , Receptor de Angiotensina Tipo 1/metabolismo , Tetrazoles/uso terapéutico , Bloqueadores del Receptor Tipo 1 de Angiotensina II/administración & dosificación , Bloqueadores del Receptor Tipo 1 de Angiotensina II/química , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Animales , Bencimidazoles/administración & dosificación , Bencimidazoles/química , Bencimidazoles/metabolismo , Compuestos de Bifenilo , Línea Celular Tumoral , Quitosano/química , Quitosano/metabolismo , ADN/genética , ADN/uso terapéutico , Sistemas de Liberación de Medicamentos , Técnicas de Transferencia de Gen , Terapia Genética , Humanos , Masculino , Ratones Endogámicos BALB C , Neoplasias/irrigación sanguínea , Neoplasias/genética , Neoplasias/metabolismo , Neovascularización Patológica/genética , Polietileneimina/química , Polietileneimina/metabolismo , Tetrazoles/administración & dosificación , Tetrazoles/química , Tetrazoles/metabolismo
12.
Exp Mol Pathol ; 96(3): 431-7, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24768585

RESUMEN

Mammalian target of rapamycin (mTOR) has been reported to contribute to the development of HIV-associated nephropathy (HIVAN). We hypothesized that HIV may be activating renal tissue mTOR pathway through renin angiotensin system (RAS) via Angiotensin Receptor Type II receptor (AT2R). Renal tissues of Vpr transgenic and Tg26 (HIVAN) mice displayed enhanced phosphorylation of mTOR and p70S6K. Aliskiren, a renin inhibitor attenuated phosphorylation of both mTOR and p70S6K in renal tissues of HIVAN mice. Interestingly, Angiotensin Receptor Type I (AT1R) blockade did not modulate renal tissue phosphorylation of mTOR in HIVAN mice; on the other hand, AT2R blockade attenuated renal tissue phosphorylation of mTOR in HIVAN mice. In vitro studies, both renin and Ang II displayed enhanced mouse tubular cell (MTC) phosphorylation of p70S6K in a dose dependent manner. HIV/MTC also displayed enhanced phosphorylation of both mTOR and p70S6K; interestingly this effect of HIV was further enhanced by losartan (an AT1R blocker). On the other hand, AT2R blockade attenuated HIV-induced tubular cell phosphorylation of mTOR and p70S6K, whereas, AT2R agonist enhanced phosphorylation of mTOR and p70S6K. These findings indicate that HIV stimulates mTOR pathway in HIVAN through the activation of renin angiotensin system via AT2R.


Asunto(s)
Nefropatía Asociada a SIDA/genética , Enfermedades Renales/virología , Receptor de Angiotensina Tipo 2/metabolismo , Sistema Renina-Angiotensina/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Amidas/farmacología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Bloqueadores del Receptor Tipo 2 de Angiotensina II/metabolismo , Animales , Fumaratos/farmacología , VIH , Enfermedades Renales/veterinaria , Losartán/farmacología , Ratones , Ratones Transgénicos , Fosforilación , Receptor de Angiotensina Tipo 2/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/genética
13.
Wound Repair Regen ; 21(1): 131-40, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23231670

RESUMEN

The renin-angiotensin system (RAS) plays an important role in wound repair; however, little is known pertaining to RAS expression in response to thermal injury and the combination of radiation plus burn injury (CRBI). The purpose of this study was to test the hypothesis that thermal injury modifies expression of RAS components and CRBI delayed this up-regulation of RAS. Skin from uninjured mice was compared with mice receiving local thermal injury or CRBI (injury site). Skin was analyzed for gene and protein expression of RAS components. There was an initial increase in the expression of various components of RAS following thermal injury. However, in the higher CRBI group there is an initial decrease in AT(1b) (vasoconstriction, pro-proliferative), AT(2) (vasodilation, differentiation), and Mas (vasodilation, anti-inflammatory) gene expression. This corresponded with a delay and decrease in AT(1) , AT(2) , and MAS protein expression in fibroblasts and keratinocytes. The reduction in RAS receptor positive fibroblasts and keratinocytes correlated with a reduction in collagen deposition and keratinocyte infiltration into the wounded area resulting in a delay of reepithelialization following CRBI. These data support the hypothesis that delayed wound healing observed in subjects following radiation exposure may be in part due to decreased expression of RAS.


Asunto(s)
Quemaduras/metabolismo , Colágeno/metabolismo , Traumatismos Experimentales por Radiación/metabolismo , Sistema Renina-Angiotensina , Piel/metabolismo , Cicatrización de Heridas , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Bloqueadores del Receptor Tipo 2 de Angiotensina II/metabolismo , Animales , Quemaduras/patología , Modelos Animales de Enfermedad , Femenino , Fibroblastos/metabolismo , Expresión Génica , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Traumatismos Experimentales por Radiación/patología , Piel/lesiones , Piel/patología , Regulación hacia Arriba
14.
Exp Diabetes Res ; 2012: 271028, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22829804

RESUMEN

The metabolic syndrome affects 30% of the US population with increasing prevalence. In this paper, we explore the relationship between the metabolic syndrome and the incidence and severity of cardiovascular disease in general and coronary artery disease (CAD) in particular. Furthermore, we look at the impact of metabolic syndrome on outcomes of coronary revascularization therapies including CABG, PTCA, and coronary collateral development. We also examine the association between the metabolic syndrome and its individual component pathologies and oxidative stress. Related, we explore the interaction between the main external sources of oxidative stress, cigarette smoke and air pollution, and metabolic syndrome and the effect of this interaction on CAD. We discuss the apparent lack of positive effect of antioxidants on cardiovascular outcomes in large clinical trials with emphasis on some of the limitations of these trials. Finally, we present evidence for successful use of antioxidant properties of pharmacological agents, including metformin, statins, angiotensin II type I receptor blockers (ARBs), and angiotensin II converting enzyme (ACE) inhibitors, for prevention and treatment of the cardiovascular complications of the metabolic syndrome.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Síndrome Metabólico/metabolismo , Estrés Oxidativo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Antioxidantes/metabolismo , Enfermedad de la Arteria Coronaria/metabolismo , Humanos , Resistencia a la Insulina , Metformina/farmacología , Ratones , Obesidad/metabolismo , Factores de Riesgo
15.
Microvasc Res ; 84(3): 395-8, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22750392

RESUMEN

There is a growing body of evidence that renin-angiotensin system plays a role in diabetic nephropathy. Recently, we have found that glucagon-like peptide-1 (GLP-1), one of the incretins, a gut hormone secreted from L cells in the intestine in response to food intake, inhibits advanced glycation end product-induced monocyte chemoattractant protein-1 gene expression in mesangial cells thorugh the interaction with the receptor of GLP-1. However, effects of GLP-1 on angiotensin II-exposed mesangial cells are unknown. This study investigated whether and how GLP-1 blocked the angiotensin II-induced mesangial cell damage in vitro. GLP-1 completely blocked the angiotensin II-induced superoxide generation, NF-κB activation, up-regulation of mRNA levels of intercellular adhesion molecule-1 and plasminogen activator inhibitor-1 in mesangial cells, all of which were prevented by the treatments with H-89, an inhibitor of protein kinase A. The present results demonstrated for the first time that GLP-1 blocked the angiotensin II-induced mesangial cell injury by inhibiting superoxide-mediated NF-κB activation via protein kinase C pathway. Our present study suggests that strategies to enhance the biological actions of GLP-1 may be a promising strategy for the treatment of diabetic nephropathy.


Asunto(s)
Angiotensina II/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Regulación Enzimológica de la Expresión Génica , Péptido 1 Similar al Glucagón/fisiología , Células Mesangiales/citología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Nefropatías Diabéticas/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Hormonas/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/biosíntesis , Isoquinolinas/farmacología , FN-kappa B/metabolismo , Inhibidor 1 de Activador Plasminogénico/biosíntesis , ARN Mensajero/metabolismo , Sulfonamidas/farmacología , Regulación hacia Arriba
16.
Drug Metab Dispos ; 40(9): 1744-56, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22665271

RESUMEN

Interindividual variability in activity of uptake transporters is evident in vivo, yet limited data exist in vitro, confounding in vitro-in vivo extrapolation. The uptake kinetics of seven organic anion-transporting polypeptide substrates was investigated over a concentration range in plated cryopreserved human hepatocytes. Active uptake clearance (CL(active, u)), bidirectional passive diffusion (P(diff)), intracellular binding, and metabolism were estimated for bosentan, pitavastatin, pravastatin, repaglinide, rosuvastatin, telmisartan, and valsartan in HU4122 donor using a mechanistic two-compartment model in Matlab. Full uptake kinetics of rosuvastatin and repaglinide were also characterized in two additional donors, whereas for the remaining drugs CL(active, u) was estimated at a single concentration. The unbound affinity constant (K(m, u)) and P(diff) values were consistent across donors, whereas V(max) was on average up to 2.8-fold greater in donor HU4122. Consistency in K(m, u) values allowed extrapolation of single concentration uptake activity data and assessment of interindividual variability in CL(active) across donors. The maximal contribution of active transport to total uptake differed among donors, for example, 85 to 96% and 68 to 87% for rosuvastatin and repaglinide, respectively; however, in all cases the active process was the major contributor. In vitro-in vivo extrapolation indicated a general underprediction of hepatic intrinsic clearance, an average empirical scaling factor of 17.1 was estimated on the basis of seven drugs investigated in three hepatocyte donors, and donor-specific differences in empirical factors are discussed. Uptake K(m, u) and CL(active, u) were on average 4.3- and 7.1-fold lower in human hepatocytes compared with our previously published rat data. A strategy for the use of rat uptake data to facilitate the experimental design in human hepatocytes is discussed.


Asunto(s)
Hepatocitos/metabolismo , Modelos Biológicos , Transportadores de Anión Orgánico/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Antihipertensivos/metabolismo , Antihipertensivos/farmacología , Bencimidazoles/metabolismo , Bencimidazoles/farmacología , Benzoatos/metabolismo , Benzoatos/farmacología , Transporte Biológico , Bosentán , Carbamatos/metabolismo , Carbamatos/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Fluorobencenos/metabolismo , Fluorobencenos/farmacología , Hepatocitos/efectos de los fármacos , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Hipoglucemiantes/metabolismo , Hipoglucemiantes/farmacología , Cinética , Transportadores de Anión Orgánico/efectos de los fármacos , Piperidinas/metabolismo , Piperidinas/farmacología , Pravastatina/metabolismo , Pravastatina/farmacología , Pirimidinas/metabolismo , Pirimidinas/farmacología , Quinolinas/metabolismo , Quinolinas/farmacología , Ratas , Rosuvastatina Cálcica , Especificidad de la Especie , Sulfonamidas/metabolismo , Sulfonamidas/farmacología , Telmisartán , Tetrazoles/metabolismo , Tetrazoles/farmacología , Valina/análogos & derivados , Valina/metabolismo , Valina/farmacología , Valsartán
17.
J Am Soc Nephrol ; 23(3): 421-8, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22193384

RESUMEN

Signaling through both angiotensin AT1 receptors (AT1R) and dopamine D1 receptors (D1R) modulates renal sodium excretion and arterial BP. AT1R and D1R form heterodimers, but whether treatment with AT1R antagonists functionally modifies D1R via allosterism is unknown. In this study, the AT1R antagonist losartan strengthened the interaction between AT1R and D1R and increased expression of D1R on the plasma membrane in vitro. In rat proximal tubule cells that express endogenous AT1R and D1R, losartan increased cAMP generation. Losartan increased cAMP in HEK 293a cells transfected with both AT1R and D1R, but it did not increase cAMP in cells transfected with either receptor alone, suggesting that losartan induces D1R activation. Furthermore, losartan did not increase cAMP in HEK 293a cells expressing AT1R and mutant S397/S398A D1R, which disrupts the physical interaction between AT1R and D1R. In vivo, administration of a D1R antagonist significantly attenuated the antihypertensive effect of losartan in rats with renal hypertension. Taken together, these data imply that losartan might exert its antihypertensive effect both by inhibiting AT1R signaling and by enhancing D1R signaling.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Túbulos Renales Proximales/metabolismo , Riñón/metabolismo , Losartán/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Receptores de Dopamina D1/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Coartación Aórtica/complicaciones , Benzazepinas/farmacología , Benzazepinas/uso terapéutico , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Hipertensión/tratamiento farmacológico , Hipertensión/etiología , Técnicas In Vitro , Riñón/citología , Riñón/efectos de los fármacos , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/efectos de los fármacos , Losartán/farmacología , Losartán/uso terapéutico , Masculino , Unión Proteica , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1/efectos de los fármacos , Receptores de Dopamina D1/antagonistas & inhibidores , Receptores de Dopamina D1/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
18.
Antivir Ther ; 16(5): 639-45, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21817185

RESUMEN

BACKGROUND: Hypertension is more prevalent among HIV-infected individuals than in the general population and contributes to increased cardiovascular risk. The angiotensin II receptor blocker telmisartan is also a partial peroxisome proliferator activated receptor-γ agonist with documented effects on glucose and lipid homeostasis. The aim of this study was to evaluate the antihypertensive and metabolic effects of telmisartan in hypertensive HIV-positive patients. METHODS: A total of 18 HIV-positive men treated with antiretroviral therapy and recently diagnosed with hypertension were administered 80 mg telmisartan daily. Systolic blood pressure (SBP) and diastolic blood pressure (DBP), viroimmunological and metabolic parameters, insulin resistance, C-reactive protein, microalbuminuria, cystatin C and plasma levels of interleukin-18 and endothelin-1 were measured at baseline (T0), 1 month (T1), 3 months (T3) and 6 months (T6). RESULTS: Treatment with telmisartan not only decreased SBP and DBP levels, but also improved insulin resistance and microalbuminuria by T1. Levels of triglycerides significantly decreased and high-density lipoprotein cholesterol increased at T1, whereas total and low-density lipoprotein cholesterol levels were statistically reduced at T3 and T6. Cystatin C and endothelin-1 showed a significant reduction at T1, whereas interleukin-18 decreased at both T3 and T6. CONCLUSIONS: Telmisartan was effective in reducing blood pressure and improving lipid metabolism and renal function. Reduction of endothelin-1 might be related to an endothelial protective effect. On the basis of these findings, and because of properties unrelated to blood pressure lowering, telmisartan might be the first choice antihypertensive drug for the treatment of HIV-positive patients.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Antihipertensivos/farmacología , Bencimidazoles/farmacología , Benzoatos/farmacología , Seropositividad para VIH/complicaciones , Hipertensión/tratamiento farmacológico , Adulto , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Antihipertensivos/metabolismo , Bencimidazoles/metabolismo , Benzoatos/metabolismo , Presión Sanguínea/efectos de los fármacos , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/prevención & control , Cistatina C/sangre , Cistatina C/efectos de los fármacos , Endotelina-1/sangre , Endotelina-1/efectos de los fármacos , Seropositividad para VIH/metabolismo , Humanos , Hipertensión/complicaciones , Hipertensión/metabolismo , Hipertensión/fisiopatología , Resistencia a la Insulina , Interleucina-18/sangre , Interleucina-18/metabolismo , Lipoproteínas HDL/sangre , Lipoproteínas HDL/efectos de los fármacos , Lipoproteínas LDL/sangre , Lipoproteínas LDL/efectos de los fármacos , Masculino , Persona de Mediana Edad , PPAR gamma/agonistas , PPAR gamma/efectos de los fármacos , Factores de Riesgo , Telmisartán , Triglicéridos/sangre , Triglicéridos/metabolismo
19.
Pharmacology ; 88(1-2): 1-9, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21709429

RESUMEN

The present study was designed to investigate the effects of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors (atorvastatin, pravastatin, simvastatin) on the pharmacokinetics of losartan and its active metabolite EXP-3174 in rats. Pharmacokinetic parameters of losartan and EXP-3174 in rats were determined after oral and intravenous administration of losartan (9 mg/kg) without and with HMG-CoA reductase inhibitors (1 mg/kg). The effect of HMG-CoA reductase inhibitors on P-gp and cytochrome (CYP) 3A4 activity were also evaluated. Atorvastatin, pravastatin and simvastatin inhibited CYP3A4 activities with IC50 values of 48.0, 14.1 and 3.10 µmol/l, respectively. Simvastatin (1-10 µmol/l) enhanced the cellular uptake of rhodamine-123 in a concentration-dependent manner. The area under the plasma concentration-time curve (AUC0₋∞) and the peak plasma concentration of losartan were significantly (p < 0.05) increased by 59.6 and 45.8%, respectively, by simvastatin compared to those of control. The total body clearance (CL/F) of losartan after oral administration with simvastatin was significantly decreased (by 34.8%) compared to that of controls. Consequently, the absolute bioavailability (F) of losartan after oral administration with simvastatin was significantly increased by 59.4% compared to that of control. The metabolite-parent AUC ratio was significantly decreased by 25.7%, suggesting that metabolism of losartan was inhibited by simvastatin. In conclusion, the enhanced bioavailability of losartan might be mainly due to inhibition of P-gp in the small intestine and CYP3A subfamily-mediated metabolism of losartan in the small intestine and/or liver and to reduction of the CL/F of losartan by simvastatin.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacocinética , Anticolesterolemiantes/farmacocinética , Citocromo P-450 CYP3A/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Imidazoles/farmacocinética , Losartán/farmacocinética , Tetrazoles/farmacocinética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/análisis , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Acilcoenzima A/antagonistas & inhibidores , Administración Oral , Bloqueadores del Receptor Tipo 1 de Angiotensina II/administración & dosificación , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Anticolesterolemiantes/sangre , Anticolesterolemiantes/farmacología , Línea Celular Tumoral , Inhibidores del Citocromo P-450 CYP3A , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Femenino , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/sangre , Inhibidores de Hidroximetilglutaril-CoA Reductasas/metabolismo , Imidazoles/sangre , Imidazoles/farmacología , Inyecciones Intravenosas , Losartán/administración & dosificación , Losartán/metabolismo , Losartán/farmacología , Lovastatina/análogos & derivados , Lovastatina/farmacocinética , Lovastatina/farmacología , Masculino , Ratas , Ratas Sprague-Dawley , Rodamina 123/metabolismo , Simvastatina/sangre , Simvastatina/metabolismo , Simvastatina/farmacología , Tetrazoles/sangre , Tetrazoles/farmacología , Factores de Tiempo
20.
J Cell Mol Med ; 15(7): 1572-81, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20477906

RESUMEN

Clinical trials have shown that angiotensin II receptor blockers reduce the new onset of diabetes in hypertensives; however, the underlying mechanisms remain unknown. We investigated the effects of telmisartan on peroxisome proliferator activated receptor γ (PPAR-δ) and the adenosine monophosphate (AMP)-activated protein kinase (AMPK) pathway in cultured myotubes, as well as on the running endurance of wild-type and PPAR-δ-deficient mice. Administration of telmisartan up-regulated levels of PPAR-δ and phospho-AMPKα in cultured myotubes. However, PPAR-δ gene deficiency completely abolished the telmisartan effect on phospho-AMPKαin vitro. Chronic administration of telmisartan remarkably prevented weight gain, enhanced running endurance and post-exercise oxygen consumption, and increased slow-twitch skeletal muscle fibres in wild-type mice, but these effects were absent in PPAR-δ-deficient mice. The mechanism is involved in PPAR-δ-mediated stimulation of the AMPK pathway. Compared to the control mice, phospho-AMPKα level in skeletal muscle was up-regulated in mice treated with telmisartan. In contrast, phospho-AMPKα expression in skeletal muscle was unchanged in PPAR-δ-deficient mice treated with telmisartan. These findings highlight the ability of telmisartan to improve skeletal muscle function, and they implicate PPAR-δ as a potential therapeutic target for the prevention of type 2 diabetes.


Asunto(s)
Adenilato Quinasa/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Bencimidazoles/metabolismo , Benzoatos/metabolismo , Músculo Esquelético/fisiología , PPAR delta/metabolismo , Resistencia Física/fisiología , Carrera/fisiología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Animales , Bencimidazoles/uso terapéutico , Benzoatos/uso terapéutico , Células Cultivadas , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/citología , Consumo de Oxígeno/fisiología , PPAR delta/genética , Telmisartán
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA