Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Antimicrob Chemother ; 73(1): 109-117, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-29029095

RESUMEN

OBJECTIVES: The increasing prevalence of mutations in HIV-1 reverse transcriptase (RT) that confer resistance to existing NRTIs and NNRTIs underscores the need to develop RT inhibitors with novel mode-of-inhibition and distinct resistance profiles. METHODS: Biochemical assays were employed to identify inhibitors of RT activity and characterize their mode of inhibition. The antiviral activity of the inhibitors was assessed by cell-based assays using laboratory HIV-1 isolates and MT4 cells. RT variants were purified via avidin affinity columns. RESULTS: Compound A displayed equal or greater potency against many common NNRTI-resistant RTs (K103N and Y181C RTs) relative to WT RT. Despite possessing certain NNRTI-like properties, such as being unable to inhibit an engineered variant of RT lacking an NNRTI-binding pocket, we found that compound A was dependent on Mg2+ for binding to RT. Optimization of compound A led to more potent analogues, which retained similar activities against WT and K103N mutant viruses with submicromolar potency in a cell-based assay. One of the analogues, compound G, was crystallized in complex with RT and the structure was determined at 2.6 Å resolution. The structure indicated that compound G simultaneously interacts with the active site (Asp186), the highly conserved primer grip region (Leu234 and Trp229) and the NNRTI-binding pocket (Tyr188). CONCLUSIONS: These findings reveal a novel class of RT bifunctional inhibitors that are not sensitive to the most common RT mutations, which can be further developed to address the deficiency of current RT inhibitors.


Asunto(s)
Fármacos Anti-VIH/farmacología , Farmacorresistencia Viral/genética , Transcriptasa Inversa del VIH/antagonistas & inhibidores , VIH-1/efectos de los fármacos , Inhibidores de la Transcriptasa Inversa/farmacología , Sitios de Unión/genética , Dominio Catalítico/efectos de los fármacos , Transcriptasa Inversa del VIH/genética , Humanos
2.
Proc Natl Acad Sci U S A ; 111(10): 3721-6, 2014 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-24567384

RESUMEN

Disease associated with Clostridium difficile infection is caused by the actions of the homologous toxins TcdA and TcdB on colonic epithelial cells. Binding to target cells triggers toxin internalization into acidified vesicles, whereupon cryptic segments from within the 1,050-aa translocation domain unfurl and insert into the bounding membrane, creating a transmembrane passageway to the cytosol. Our current understanding of the mechanisms underlying pore formation and the subsequent translocation of the upstream cytotoxic domain to the cytosol is limited by the lack of information available regarding the identity and architecture of the transmembrane pore. Here, through systematic perturbation of conserved sites within predicted membrane-insertion elements of the translocation domain, we uncovered highly sensitive residues--clustered between amino acids 1,035 and 1,107--that when individually mutated, reduced cellular toxicity by as much as >1,000-fold. We demonstrate that defective variants are defined by impaired pore formation in planar lipid bilayers and biological membranes, resulting in an inability to intoxicate cells through either apoptotic or necrotic pathways. These findings along with the unexpected similarities uncovered between the pore-forming "hotspots" of TcdB and the well-characterized α-helical diphtheria toxin translocation domain provide insights into the structure and mechanism of formation of the translocation pore for this important class of pathogenic toxins.


Asunto(s)
Toxinas Bacterianas/genética , Toxinas Bacterianas/toxicidad , Clostridioides difficile/patogenicidad , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/toxicidad , Secuencia de Aminoácidos , Toxinas Bacterianas/metabolismo , Clostridioides difficile/genética , Fluorescencia , Ensayos Analíticos de Alto Rendimiento , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis , Mutación/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Estructura Terciaria de Proteína/genética , Radioisótopos de Rubidio/metabolismo
3.
J Biol Chem ; 290(3): 1474-84, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25355312

RESUMEN

The pyrophosphate mimic and broad spectrum antiviral phosphonoformic acid (PFA, foscarnet) was shown to freeze the pre-translocational state of the reverse transcriptase (RT) complex of the human immunodeficiency virus type 1 (HIV-1). However, PFA lacks a specificity domain, which is seen as a major reason for toxic side effects associated with the clinical use of this drug. Here, we studied the mechanism of inhibition of HIV-1 RT by the 4-chlorophenylhydrazone of mesoxalic acid (CPHM) and demonstrate that this compound also blocks RT translocation. Hot spots for inhibition with PFA or CPHM occur at template positions with a bias toward pre-translocation. Mutations at active site residue Asp-185 compromise binding of both compounds. Moreover, divalent metal ions are required for the formation of ternary complexes with either of the two compounds. However, CPHM contains both an anchor domain that likely interacts with the catalytic metal ions and a specificity domain. Thus, although the inhibitor binding sites may partly overlap, they are not identical. The K65R mutation in HIV-1 RT, which reduces affinity to PFA, increases affinity to CPHM. Details with respect to the binding sites of the two inhibitors are provided on the basis of mutagenesis studies, structure-activity relationship analyses with newly designed CPHM derivatives, and in silico docking experiments. Together, these findings validate the pre-translocated complex of HIV-1 RT as a specific target for the development of novel classes of RT inhibitors.


Asunto(s)
Transcriptasa Inversa del VIH/antagonistas & inhibidores , VIH-1/enzimología , Hidrazonas/química , Malonatos/química , Inhibidores de la Transcriptasa Inversa/química , Antirretrovirales/química , Catálisis , Dominio Catalítico , Evaluación Preclínica de Medicamentos , Iones , Metales/química , Modelos Moleculares , Mutagénesis , Mutación , Unión Proteica , Multimerización de Proteína , Ribonucleasa H/química , Relación Estructura-Actividad
4.
Mol Pharm ; 12(8): 2962-71, 2015 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-26103531

RESUMEN

Platforms enabling targeted delivery of proteins into cells are needed to fully realize the potential of protein-based therapeutics with intracellular sites-of-action. Bacterial toxins are attractive systems to consider as templates for designing protein transduction systems as they naturally bind and enter specific cells with high efficiency. Here we investigated the capacity of diphtheria toxin to function as an intracellular protein delivery vector. We report that diphtheria toxin delivers an impressive array of passenger proteins spanning a range of sizes, structures, and stabilities into cells in a manner that indicates that they are "invisible" to the translocation machinery. Further, we show that α-amylase delivered into cells by a detoxified diphtheria toxin chimera digests intracellular glycogen in live cells, providing evidence that delivered cargo is folded, active, and abundant. The efficiency and versatility of diphtheria toxin over existing systems open numerous possibilities for intracellular delivery of bioactive proteins.


Asunto(s)
Toxina Diftérica/metabolismo , Sistemas de Liberación de Medicamentos , Glucógeno/metabolismo , Fragmentos de Péptidos/metabolismo , alfa-Amilasas/química , alfa-Amilasas/metabolismo , Rastreo Diferencial de Calorimetría , Células HEK293 , Humanos , Pliegue de Proteína
5.
J Med Chem ; 67(4): 2349-2368, 2024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38299539

RESUMEN

ATR is a key kinase in the DNA-damage response (DDR) that is synthetic lethal with several other DDR proteins, making it an attractive target for the treatment of genetically selected solid tumors. Herein we describe the discovery of a novel ATR inhibitor guided by a pharmacophore model to position a key hydrogen bond. Optimization was driven by potency and selectivity over the related kinase mTOR, resulting in the identification of camonsertib (RP-3500) with high potency and excellent ADME properties. Preclinical evaluation focused on the impact of camonsertib on myelosuppression, and an exploration of intermittent dosing schedules to allow recovery of the erythroid compartment and mitigate anemia. Camonsertib is currently undergoing clinical evaluation both as a single agent and in combination with talazoparib, olaparib, niraparib, lunresertib, or gemcitabine (NCT04497116, NCT04972110, NCT04855656). A preliminary recommended phase 2 dose for monotherapy was identified as 160 mg QD given 3 days/week.


Asunto(s)
Neoplasias , Humanos , Proteínas de la Ataxia Telangiectasia Mutada , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Gemcitabina
6.
Microbiol Spectr ; 11(3): e0029623, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37098898

RESUMEN

Biosynthesis of the Pel exopolysaccharide in Pseudomonas aeruginosa requires all seven genes of the pelABCDEFG operon. The periplasmic modification enzyme PelA contains a C-terminal deacetylase domain that is necessary for Pel-dependent biofilm formation. Herein, we show that extracellular Pel is not produced by a P. aeruginosa PelA deacetylase mutant. This positions PelA deacetylase activity as an attractive target to prevent Pel-dependent biofilm formation. Using a high-throughput screen (n = 69,360), we identified 56 compounds that potentially inhibit PelA esterase activity, the first enzymatic step in the deacetylase reaction. A secondary biofilm inhibition assay identified methyl 2-(2-pyridinylmethylene) hydrazinecarbodithioate (SK-017154-O) as a specific Pel-dependent biofilm inhibitor. Structure-activity relationship studies identified the thiocarbazate as a necessary functional group and that the pyridyl ring could be replaced with a phenyl substituent (compound 1). Both SK-017154-O and compound 1 inhibit Pel-dependent biofilm formation in Bacillus cereus ATCC 10987, which has a predicted extracellular PelA deacetylase in its pel operon. Michaelis-Menten kinetics determined SK-017154-O to be a noncompetitive inhibitor of PelA, while compound 1 did not directly inhibit PelA esterase activity. Cytotoxicity assays using human lung fibroblast cells showed that compound 1 is less cytotoxic than SK-017154-O. This work provides proof of concept that biofilm exopolysaccharide modification enzymes are important for biofilm formation and can serve as useful antibiofilm targets. IMPORTANCE Present in more than 500 diverse Gram-negative and 900 Gram-positive organisms, the Pel polysaccharide is one of the most phylogenetically widespread biofilm matrix determinants found to date. Partial de-N-acetylation of this α-1,4 linked N-acetylgalactosamine polymer by the carbohydrate modification enzyme PelA is required for Pel-dependent biofilm formation in Pseudomonas aeruginosa and Bacillus cereus. Given this and our observation that extracellular Pel is not produced by a P. aeruginosa PelA deactylase mutant, we developed an enzyme-based high-throughput screen and identified methyl 2-(2-pyridinylmethylene) hydrazinecarbodithioate (SK-017154-O) and its phenyl derivative as specific Pel-dependent biofilm inhibitors. Michaelis-Menten kinetics revealed SK-017154-O is a noncompetitive inhibitor and that its noncytotoxic, phenyl derivative does not directly inhibit P. aeruginosa PelA esterase activity. We provide proof of concept that exopolysaccharide modification enzymes can be targeted with small molecule inhibitors to block Pel-dependent biofilm development in both Gram-negative and Gram-positive bacteria.


Asunto(s)
Polisacáridos Bacterianos , Pseudomonas aeruginosa , Humanos , Pseudomonas aeruginosa/genética , Biopelículas , Periplasma , Esterasas , Proteínas Bacterianas/genética
7.
J Biol Chem ; 286(34): 29575-83, 2011 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-21737446

RESUMEN

The rapid emergence and the prevalence of resistance mutations in HIV-1 reverse transcriptase (RT) underscore the need to identify RT inhibitors with novel binding modes and mechanisms of inhibition. Recently, two structurally distinct inhibitors, phosphonoformic acid (foscarnet) and INDOPY-1 were shown to disrupt the translocational equilibrium of RT during polymerization through trapping of the enzyme in the pre- and the post-translocation states, respectively. Here, we show that foscarnet and INDOPY-1 additionally display a shared novel inhibitory preference with respect to substrate primer identity. In RT-catalyzed reactions using RNA-primed substrates, translocation inhibitors were markedly less potent at blocking DNA polymerization than in equivalent DNA-primed assays; i.e. the inverse pattern observed with marketed non-nucleoside inhibitors that bind the allosteric pocket of RT. This potency profile was shown to correspond with reduced binding on RNA·DNA primer/template substrates versus DNA·DNA substrates. Furthermore, using site-specific footprinting with chimeric RNA·DNA primers, we demonstrate that the negative impact of the RNA primer on translocation inhibitor potency is overcome after 18 deoxyribonucleotide incorporations, where RT transitions primarily into polymerization-competent binding mode. In addition to providing a simple means to identify similarly acting translocation inhibitors, these findings suggest a broader role for the primer-influenced binding mode on RT translocation equilibrium and inhibitor sensitivity.


Asunto(s)
Cartilla de ADN/química , ADN Viral/química , Transcriptasa Inversa del VIH/antagonistas & inhibidores , Transcriptasa Inversa del VIH/química , VIH-1/enzimología , Indoles/química , Nitrilos/química , Piridonas/química , ARN Viral/química , Sitio Alostérico , Catálisis , Cartilla de ADN/metabolismo , ADN Viral/biosíntesis , Transcriptasa Inversa del VIH/metabolismo , Indoles/metabolismo , Nitrilos/metabolismo , Piridonas/metabolismo , ARN Viral/biosíntesis , Transcripción Reversa/fisiología
8.
Am J Respir Cell Mol Biol ; 39(3): 324-36, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18403781

RESUMEN

Lung fibrosis is characterized by excessive accumulation of extracellular matrix components leading to progressive airflow limitation. Distinct profibrotic pathways converge on the activation of transforming growth factor-beta (TGF-beta), a central growth factor implicated in most fibroproliferative diseases. Recently, enforced expression of bioactive human TGF-beta1 (hTGF-beta1) in lungs of transgenic mice was shown to recapitulate several key pathophysiologies observed in fibrotic disorders of the lung, including cellular inflammation, tissue fibrosis, and myofibroblast hyperplasia. Inducible expression of hTGF-beta1 in this system provided a unique opportunity to characterize TGF-beta-driven mechanisms that precede and/or follow the onset of inflammation and fibrosis. Using gene expression profiling in lungs, we demonstrate temporal activation of key genetic programs regulating cell movement and invasiveness, inflammation, organ remodeling, and fibrosis. Consistent with our gene expression data, multiple soluble mediators associated with inflammation and tissue remodeling were markedly elevated in the bronchoalveolar lavage fluid of mice expressing hTGF-beta1. We observe significant TGF-beta1-driven infiltration of F4/80+ mononuclear cells producing bioactive arginase, a marker of alternatively activated macrophages. Finally, we identified a common "fibrosis" gene signature when comparing our findings with published data derived from preclinical and clinical studies.


Asunto(s)
Quimiocinas/genética , Fibrosis Pulmonar/genética , Factor de Crecimiento Transformador beta1/fisiología , Enfermedad Aguda , Animales , Bleomicina/farmacología , Líquido del Lavado Bronquioalveolar , Quimiotaxis de Leucocito , Doxiciclina/administración & dosificación , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Pulmón/metabolismo , Activación de Macrófagos , Ratones , Ratones Transgénicos , Fenotipo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/patología , Factor de Crecimiento Transformador beta1/genética
9.
Mol Endocrinol ; 21(4): 797-816, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17299137

RESUMEN

The basis for the differential repressive effects of antiestrogens on transactivation by estrogen receptor-alpha (ERalpha) remains incompletely understood. Here, we show that the full antiestrogen ICI182,780 and, to a lesser extent, the selective ER modulator raloxifene (Ral), induce accumulation of exogenous ERalpha in a poorly soluble fraction in transiently transfected HepG2 or stably transfected MDA-MB231 cells and of endogenous receptor in MCF7 cells. ERalpha remained nuclear in HepG2 cells treated with either compound. Replacement of selected hydrophobic residues of ERalpha ligand-binding domain helix 12 (H12) enhanced receptor solubility in the presence of ICI182,780 or Ral. These mutations also increased transcriptional activity with Ral or ICI182,780 on reporter genes or on the endogenous estrogen target gene TFF1 in a manner requiring the integrity of the N-terminal AF-1 domain. The antiestrogen-specific effects of single mutations suggest that they affect receptor function by mechanisms other than a simple decrease in hydrophobicity of H12, possibly due to relief from local steric hindrance between these residues and the antiestrogen side chains. Fluorescence anisotropy experiments indicated an enhanced regional stabilization of mutant ligand-binding domains in the presence of antiestrogens. H12 mutations also prevent the increase in bioluminescence resonance energy transfer between ERalpha monomers induced by Ral or ICI182,780 and increase intranuclear receptor mobility in correlation with transcriptional activity in the presence of these antiestrogens. Our data indicate that ICI182,780 and Ral locally alter the ERalpha ligand binding structure via specific hydrophobic residues of H12 and decrease its transcriptional activity through tighter association with an insoluble nuclear structure.


Asunto(s)
Núcleo Celular/metabolismo , Estradiol/análogos & derivados , Antagonistas de Estrógenos/farmacología , Receptor alfa de Estrógeno/efectos de los fármacos , Leucina/metabolismo , Clorhidrato de Raloxifeno/farmacología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Aminoácidos/química , Aminoácidos/genética , Aminoácidos/metabolismo , Núcleo Celular/química , Estradiol/farmacología , Receptor alfa de Estrógeno/análisis , Receptor alfa de Estrógeno/metabolismo , Fulvestrant , Humanos , Leucina/química , Leucina/genética , Datos de Secuencia Molecular , Mutación , Estructura Secundaria de Proteína , Solubilidad , Transcripción Genética/efectos de los fármacos , Factor Trefoil-1 , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/genética
10.
FASEB J ; 19(13): 1911-3, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16148026

RESUMEN

Rho GTPases are key regulators of many cellular functions, including cytoskeleton organization which is important for cell morphology and mobility, gene expression, cell cycle progression, and cytokinesis. In addition, it has recently been recognized that Rho GTPase activity is required for development of the immune system, as well as for the specialized functions of the peripheral cells that act in the immune response such as antigen presenting cells and lymphocytes. Stimulation of T lymphocytes with interleukin-2 (IL-2) induces clonal expansion of antigen-specific populations and provides a model to study cell cycle entry and cell cycle progression. We have performed gene expression analysis in a model of human T lymphocytes, which proliferate in response to IL-2. In addition to changes in genes relevant to cell cycling and to the antiapoptotic effects of IL-2, we have analyzed expression and variations of more than 300 genes involved in Rho GTPase signaling pathways. We report here that IL-2 regulates the expression of a number of proteins, which participate in the Rho GTPase pathways, including some of the GTPases themselves, GDP/GTP exchange factors, GTPase activating proteins, as well as GDIs and effectors. Our results suggest that regulation of expression of components of the Rho GTPase pathways may be an important mechanism in assembling specific signal transduction cascades that need to be active at certain times during the cell cycle. Some of our findings may also be relevant to the roles of Rho GTPases in T lymphocyte functions and proliferation.


Asunto(s)
Transducción de Señal , Linfocitos T/inmunología , Proteínas de Unión al GTP rho/metabolismo , Western Blotting , Linfocitos T CD4-Positivos/metabolismo , Ciclo Celular , Línea Celular , Proliferación Celular , Cicloheximida/farmacología , Progresión de la Enfermedad , Citometría de Flujo , Proteínas Activadoras de GTPasa/metabolismo , Regulación de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Sistema Inmunológico , Interleucina-2/metabolismo , Hibridación de Ácido Nucleico , Análisis de Secuencia por Matrices de Oligonucleótidos , Inhibidores de la Síntesis de la Proteína/farmacología , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Intercambio de Guanina Nucleótido Rho , Linfocitos T/metabolismo , Factores de Tiempo
11.
Chem Biol ; 22(2): 175-85, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-25619932

RESUMEN

Clostridium difficile causes life-threatening diarrhea through the actions of its homologous toxins TcdA and TcdB on human colonocytes. Therapeutic agents that block toxin-induced damage are urgently needed to prevent the harmful consequences of toxin action that are not addressed with current antibiotic-based treatments. Here, we developed an imaging-based phenotypic screen to identify small molecules that protected human cells from TcdB-induced cell rounding. A series of structurally diverse compounds with antitoxin activity were identified and found to act through one of a small subset of mechanisms, including direct binding and sequestration of TcdB, inhibition of endosomal maturation, and noncompetitive inhibition of the toxin glucosyltransferase activity. Distinct classes of inhibitors were used further to dissect the determinants of the toxin-mediated necrosis phenotype occurring at higher doses of toxin. These findings validate and inform novel targeting strategies for discovering small molecule agents to treat C. difficile infection.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Toxinas Bacterianas/antagonistas & inhibidores , Clostridioides difficile/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Animales , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidad , Biflavonoides/química , Biflavonoides/metabolismo , Catequina/análogos & derivados , Catequina/química , Catequina/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Colatos/química , Colatos/metabolismo , Ácido Gálico/análogos & derivados , Ácido Gálico/química , Ácido Gálico/metabolismo , Humanos , Cinética , Necrosis , Floretina/química , Floretina/metabolismo , Unión Proteica , Bibliotecas de Moléculas Pequeñas/metabolismo , Células Vero
12.
J Biomol Screen ; 16(5): 518-24, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21474837

RESUMEN

The authors have devised a continuous fluorescence-based assay to measure HIV reverse transcriptase (RT) polymerase activity for both high-throughput screening (HTS) and mechanistic characterization of inhibitors. The designed substrate is composed of a recessed DNA primer annealed to a DNA template that is labeled at the 5'-terminus with a donor fluorophore (AlexaFluor 488). RT-catalyzed incorporation of an acceptor-labeled deoxyuridine (dUTP-AlexaFluor 555) at the 3'-terminus of the fully extended DNA primer juxtaposes donor and acceptor fluorophores, resulting in robust fluorescence resonance energy transfer that can be monitored kinetically in real time. The assay is sensitive, permitting the use of low enzyme concentrations (<0.5 nM), and can be miniaturized for use in 384-well HTS mode. The authors further show that this assay is capable of evaluating inhibitor mechanism of action by confirming the binding mechanism of a set of nonnucleoside RT inhibitors. Given the versatility and the lack of requirement for costly platforms or radioactivity, this assay may serve to accelerate and streamline the discovery and characterization process for future antiviral agents.


Asunto(s)
ADN Polimerasa Dirigida por ADN/metabolismo , Descubrimiento de Drogas/métodos , Transcriptasa Inversa del VIH/antagonistas & inhibidores , Transcriptasa Inversa del VIH/metabolismo , Ensayos Analíticos de Alto Rendimiento , Inhibidores de la Síntesis del Ácido Nucleico , Inhibidores de la Transcriptasa Inversa/farmacología , Activación Enzimática/efectos de los fármacos , Transferencia Resonante de Energía de Fluorescencia , Cinética , Inhibidores de la Transcriptasa Inversa/química
13.
J Renin Angiotensin Aldosterone Syst ; 12(3): 133-45, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21393355

RESUMEN

INTRODUCTION: The hypertensive double-transgenic (dTG) rat strain, expressing human renin and angiotensinogen, develops severe hypertension and organ damage and 50% of individuals die by 7 weeks of age. Here, we characterise a variation of this model in which animals present stable hypertension. MATERIALS AND METHODS: The effect of renin-angiotensin system blockers on blood pressure was determined with adult dTG rats treated with enalapril from 3 to 12 weeks of age. Tissue expression levels of renin and angiotensinogen were determined in dTG rats and rhesus monkeys by quantitative PCR. RESULTS: Upon withdrawal from enalapril, mean arterial pressure (MAP) rose to 160-180 mmHg, with 95% of the female dTG rats surviving for 6 to 12 months, In Sprague-Dawley (SD) rats and rhesus monkeys, renin mRNA was absent or weakly expressed in most tissues, except for the kidneys and adrenals. In dTG rats, human renin expression was high in many additional tissues. The expression of human angiotensinogen in dTG rats followed a similar tissue pattern to SD and rhesus monkey angiotensinogen. Oral dosing of aliskiren, enalapril or losartan provided a similar maximal reduction in MAP and duration of efficacy in telemetrised dTG rats. CONCLUSIONS: Enalapril-pretreated dTG rats are suitable for long-term MAP monitoring and sequential evaluation of human renin inhibitors.


Asunto(s)
Enalapril/farmacología , Enalapril/uso terapéutico , Hipertensión/tratamiento farmacológico , Renina/antagonistas & inhibidores , Amidas/administración & dosificación , Amidas/farmacología , Amidas/uso terapéutico , Angiotensinógeno/genética , Angiotensinógeno/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Modelos Animales de Enfermedad , Enalapril/administración & dosificación , Femenino , Fumaratos/administración & dosificación , Fumaratos/farmacología , Fumaratos/uso terapéutico , Regulación de la Expresión Génica/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Hipertensión/fisiopatología , Macaca mulatta , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Renina/sangre , Renina/genética , Distribución Tisular/efectos de los fármacos
14.
Mol Pharmacol ; 70(2): 579-88, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16679488

RESUMEN

The bulky side chains of antiestrogens hinder folding of the ligand binding domain (LBD) of estrogen receptors (ERs) into a transcriptionally active conformation. The presence of a tertiary amine in the side chain of raloxifene, which interacts with a negatively charged residue in helix H3 of the ER LBD [Asp351 in human (h)ERalpha], is important for antiestrogenicity in animal and cellular models. To better understand the molecular basis of the differential activity of tamoxifen and raloxifene, we have examined the influence of tertiary amine substituents and of mutations at position 351 in hERalpha on the activity profiles of tamoxifen derivatives. Results obtained in several cellular model systems suggest that the degree of antagonist activity of tamoxifen derivatives does not strictly correlate with the basicity of the side chain but depends on an optimal spatial relationship between the tertiary amine of these antiestrogens and the negative charge at position 351. Although altering the position of the negative charge at residue 351 (mutation D351E) had little effect on transcriptional activity in the presence of tamoxifen, it drastically increased the partial agonist activity of a tamoxifen derivative with improved antagonist activity as well as that of raloxifene. Our results suggest that contrary to raloxifene, tamoxifen and most of its derivatives do not interact with Asp351 in an optimal manner, although this can be improved by modifying tertiary amine substituents.


Asunto(s)
Antagonistas de Estrógenos/farmacología , Receptor alfa de Estrógeno/química , Clorhidrato de Raloxifeno/farmacología , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Tamoxifeno/farmacología , Ácido Aspártico , Línea Celular , Receptor alfa de Estrógeno/agonistas , Receptor alfa de Estrógeno/análisis , Receptor alfa de Estrógeno/metabolismo , Humanos , Clorhidrato de Raloxifeno/metabolismo , Relación Estructura-Actividad , Tamoxifeno/metabolismo
15.
Mol Pharmacol ; 68(6): 1852-62, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16186250

RESUMEN

Histone deacetylase inhibitors (HDACi), which have emerged as a new class of anticancer agents, act by modulating expression of genes controlling apoptosis or cell proliferation. Here, we compared the effect of HDACi on transcriptional activation by estrogen or glucocorticoid receptors (ER and GR, respectively), two members of the steroid receptor family with cell growth regulatory properties. Like other transcription factors, steroid receptors modulate histone acetylation on target promoters. Using episomal reporter vectors containing minimal promoters to avoid promoter-specific effects, we observed that long-term (24-h) incubation with HDACi strongly stimulated GR-dependent but markedly repressed ER-dependent signaling in ER+/GR+ human endometrial carcinoma Ishikawa cells. These effects were reproduced on endogenous target genes and required incubation periods with HDACi substantially longer than necessary to increase global histone acetylation. Repression of estrogen signaling was due to direct inhibition of transcription from multiple ERalpha promoters and correlated with decreased histone acetylation of these promoters. In contrast, the strong HDACi stimulation of GR-dependent gene regulation was not accounted for by increased GR expression, but it was mimicked by overexpression of the histone acetyltransferase complex component transcriptional intermediary factor 2. Together, our results demonstrate striking and opposite effects of HDACi on ER and GR signaling that involve regulatory events independent of histone hyperacetylation on receptor target promoters.


Asunto(s)
Neoplasias Endometriales/patología , Inhibidores de Histona Desacetilasas , Receptores de Estrógenos/metabolismo , Receptores de Glucocorticoides/metabolismo , Transducción de Señal , Acetilación , Antineoplásicos/farmacología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Histonas/metabolismo , Humanos , Regiones Promotoras Genéticas , Activación Transcripcional/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA