Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
HIV Med ; 25(2): 212-222, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37775947

RESUMEN

OBJECTIVES: The main objective of this analysis was to evaluate the impact of pre-existing drug resistance by next-generation sequencing (NGS) on the risk of treatment failure (TF) of first-line regimens in participants enrolled in the START study. METHODS: Stored plasma from participants with entry HIV RNA >1000 copies/mL were analysed using NGS (llumina MiSeq). Pre-existing drug resistance was defined using the mutations considered by the Stanford HIV Drug Resistance Database (HIVDB v8.6) to calculate the genotypic susceptibility score (GSS, estimating the number of active drugs) for the first-line regimen at the detection threshold windows of >20%, >5%, and >2% of the viral population. Survival analysis was conducted to evaluate the association between the GSS and risk of TF (viral load >200 copies/mL plus treatment change). RESULTS: Baseline NGS data were available for 1380 antiretroviral therapy (ART)-naïve participants enrolled over 2009-2013. First-line ART included a non-nucleoside reverse transcriptase inhibitor (NNRTI) in 976 (71%), a boosted protease inhibitor in 297 (22%), or an integrase strand transfer inhibitor in 107 (8%). The proportions of participants with GSS <3 were 7% for >20%, 10% for >5%, and 17% for the >2% thresholds, respectively. The adjusted hazard ratio of TF associated with a GSS of 0-2.75 versus 3 in the subset of participants with mutations detected at the >2% threshold was 1.66 (95% confidence interval 1.01-2.74; p = 0.05) and 2.32 (95% confidence interval 1.32-4.09; p = 0.003) after restricting the analysis to participants who started an NNRTI-based regimen. CONCLUSIONS: Up to 17% of participants initiated ART with a GSS <3 on the basis of NGS data. Minority variants were predictive of TF, especially for participants starting NNRTI-based regimens.


Asunto(s)
Fármacos Anti-VIH , Infecciones por VIH , Seropositividad para VIH , VIH-1 , Humanos , Infecciones por VIH/epidemiología , VIH-1/genética , Fármacos Anti-VIH/farmacología , Fármacos Anti-VIH/uso terapéutico , Inhibidores de la Transcriptasa Inversa/uso terapéutico , Seropositividad para VIH/tratamiento farmacológico , Secuenciación de Nucleótidos de Alto Rendimiento , Carga Viral , Farmacorresistencia Viral/genética
2.
Proc Natl Acad Sci U S A ; 109(1): 143-8, 2012 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-22187462

RESUMEN

We report the three-dimensional structure of a ß-catenin armadillo repeat in complex with the liver receptor homolog-1 (LRH-1) ligand binding domain at 2.8 Å resolution as the first structure of ß-catenin in complex with any nuclear receptor. The surface of ß-catenin that binds LRH-1 partly overlaps defined contact sites for peptide segments of ß-catenin partners, including T-cell factor-4. The surface of LRH-1 that engages ß-catenin is comprised of helices 1, 9, and 10 and is distinct from known interaction surfaces of LRH-1, including corepressor and coactivator binding sites. Targeted mutagenesis of amino acids forming both sides of the LRH-1/ß-catenin interface reveals that they are essential for stable interactions between these proteins in solution. The LRH-1 binding site in ß-catenin is also required for association with androgen receptor, providing evidence that the observed LRH-1/ß-catenin interaction may be prototypic.


Asunto(s)
Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/metabolismo , beta Catenina/química , beta Catenina/metabolismo , Análisis Mutacional de ADN , Pruebas de Enzimas , Humanos , Luciferasas/metabolismo , Modelos Moleculares , Mutación Puntual , Unión Proteica , Estructura Terciaria de Proteína , Receptores Androgénicos/metabolismo , Relación Estructura-Actividad
3.
J Gen Virol ; 95(Pt 1): 142-152, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24136366

RESUMEN

The early region 1A (E1A) of human adenovirus types 2 and 5 is differentially spliced to yield five distinct mRNAs that encode different proteins. The smallest E1A RNA transcript encodes a 55 residue (55R) protein that shares only 28 amino acid residues with the other E1A proteins. Even though it is the most abundant E1A transcript at late times post-infection, little is known about the functions of this E1A isoform. In this study, we show that the E1A 55R protein interacts with, and modulates the activity of the unliganded thyroid hormone receptor (TR). We demonstrate that E1A 55R contains a signature motif known as the CoRNR box that confers interaction with the unliganded TR; this motif was originally identified in cellular corepressors. Using a system reconstituted in the yeast Saccharomyces cerevisiae, which lack endogenous TR and TR coregulators, we show that E1A 55R nonetheless differs from cellular corepressors as it functions as a strong co-activator of TR-dependent transcription and that it possesses an intrinsic transcriptional activation domain. These data indicate that the E1A 55R protein functions as a transcriptional regulator.


Asunto(s)
Proteínas E1A de Adenovirus/química , Proteínas E1A de Adenovirus/metabolismo , Infecciones por Adenovirus Humanos/genética , Adenovirus Humanos/metabolismo , Regiones Promotoras Genéticas , Receptores de Hormona Tiroidea/genética , Transactivadores/metabolismo , Proteínas E1A de Adenovirus/genética , Infecciones por Adenovirus Humanos/metabolismo , Infecciones por Adenovirus Humanos/virología , Adenovirus Humanos/química , Adenovirus Humanos/genética , Línea Celular Tumoral , Regulación de la Expresión Génica , Humanos , Unión Proteica , Estructura Terciaria de Proteína , Transactivadores/química , Transactivadores/genética , Activación Transcripcional
4.
J Biol Chem ; 287(33): 28169-79, 2012 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-22584573

RESUMEN

The recent discovery that peroxisome proliferator-activated receptor γ (PPARγ) targeted anti-diabetic drugs function by inhibiting Cdk5-mediated phosphorylation of the receptor has provided a new viewpoint to evaluate and perhaps develop improved insulin-sensitizing agents. Herein we report the development of a novel thiazolidinedione that retains similar anti-diabetic efficacy as rosiglitazone in mice yet does not elicit weight gain or edema, common side effects associated with full PPARγ activation. Further characterization of this compound shows GQ-16 to be an effective inhibitor of Cdk5-mediated phosphorylation of PPARγ. The structure of GQ-16 bound to PPARγ demonstrates that the compound utilizes a binding mode distinct from other reported PPARγ ligands, although it does share some structural features with other partial agonists, such as MRL-24 and PA-082, that have similarly been reported to dissociate insulin sensitization from weight gain. Hydrogen/deuterium exchange studies reveal that GQ-16 strongly stabilizes the ß-sheet region of the receptor, presumably explaining the compound's efficacy in inhibiting Cdk5-mediated phosphorylation of Ser-273. Molecular dynamics simulations suggest that the partial agonist activity of GQ-16 results from the compound's weak ability to stabilize helix 12 in its active conformation. Our results suggest that the emerging model, whereby "ideal" PPARγ-based therapeutics stabilize the ß-sheet/Ser-273 region and inhibit Cdk5-mediated phosphorylation while minimally invoking adipogenesis and classical agonism, is indeed a valid framework to develop improved PPARγ modulators that retain antidiabetic actions while minimizing untoward effects.


Asunto(s)
Hipoglucemiantes/farmacología , PPAR gamma/agonistas , Tiazolidinedionas/farmacología , Aumento de Peso , Células 3T3-L1 , Animales , Quinasa 5 Dependiente de la Ciclina/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Evaluación Preclínica de Medicamentos , Humanos , Hipoglucemiantes/química , Hipoglucemiantes/farmacocinética , Ligandos , Ratones , Células 3T3 NIH , PPAR gamma/genética , PPAR gamma/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/genética , Estructura Secundaria de Proteína , Tiazolidinedionas/química , Tiazolidinedionas/farmacocinética , Células U937
5.
N Engl J Med ; 362(10): 906-16, 2010 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-20220185

RESUMEN

BACKGROUND: Dyslipidemia increases the risk of atherosclerotic cardiovascular disease and is incompletely reversed by statin therapy alone in many patients. Thyroid hormone lowers levels of serum low-density lipoprotein (LDL) cholesterol and has other potentially favorable actions on lipoprotein metabolism. Consequently, thyromimetic drugs hold promise as lipid-lowering agents if adverse effects can be avoided. METHODS: We performed a randomized, placebo-controlled, double-blind, multicenter trial to assess the safety and efficacy of the thyromimetic compound eprotirome (KB2115) in lowering the level of serum LDL cholesterol in patients with hypercholesterolemia who were already receiving simvastatin or atorvastatin. In addition to statin treatment, patients received either eprotirome (at a dose of 25, 50, or 100 microg per day) or placebo. Secondary outcomes were changes in levels of serum apolipoprotein B, triglycerides, and Lp(a) lipoprotein. Patients were monitored for potential adverse thyromimetic effects on the heart, bone, and pituitary. RESULTS: The addition of placebo or eprotirome at a dose of 25, 50, or 100 microg daily to statin treatment for 12 weeks reduced the mean level of serum LDL cholesterol from 141 mg per deciliter (3.6 mmol per liter) to 127, 113, 99, and 94 mg per deciliter (3.3, 2.9, 2.6, and 2.4 mmol per liter), respectively, (mean reduction from baseline, 7%, 22%, 28%, and 32%). Similar reductions were seen in levels of serum apolipoprotein B, triglycerides, and Lp(a) lipoprotein. Eprotirome therapy was not associated with adverse effects on the heart or bone. No change in levels of serum thyrotropin or triiodothyronine was detected, although the thyroxine level decreased in patients receiving eprotirome. CONCLUSIONS: In this 12-week trial, the thyroid hormone analogue eprotirome was associated with decreases in levels of atherogenic lipoproteins in patients receiving treatment with statins. (ClinicalTrials.gov number, NCT00593047.)


Asunto(s)
Anilidas/uso terapéutico , Dislipidemias/tratamiento farmacológico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Adulto , Anilidas/efectos adversos , LDL-Colesterol/sangre , Método Doble Ciego , Quimioterapia Combinada , Dislipidemias/sangre , Femenino , Humanos , Lipoproteínas/sangre , Masculino , Persona de Mediana Edad , Hormonas Tiroideas/sangre , Triglicéridos/sangre , Triyodotironina/análogos & derivados
6.
J Biol Chem ; 286(35): 30723-30731, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21719705

RESUMEN

Peroxisome proliferator-activated receptor γ (PPARγ) activation induces adipogenesis and also enhances lipogenesis, mitochondrial activity, and insulin sensitivity in adipocytes. Whereas some studies implicate PPARγ coactivator 1α (PGC-1α) in the mitochondrial effect, the mechanisms involved in PPARγ regulation of adipocyte mitochondrial function are not resolved. PPARγ-activating ligands (thiazolidinediones (TZDs)) are important insulin sensitizers and were recently shown to indirectly induce PGC-1ß transcription in osteoclasts. Here, we asked whether similar effects occur in adipocytes and show that TZDs also strongly induce PGC-1ß in cultured 3T3-L1 cells. This effect, however, differs from the indirect effect proposed for bone and is rapid and direct and involves PPARγ interactions with an intronic PPARγ response element cluster in the PGC-1ß locus. TZD treatment of cultured adipocytes results in up-regulation of mitochondrial marker genes, and increased mitochondrial activity and use of short interfering RNA confirms that these effects require PGC-1ß. PGC-1ß did not participate in PPARγ effects on adipogenesis or lipogenesis, and PGC-1ß knockdown did not alter insulin-responsive glucose uptake into 3T3-L1 cells. Similar effects on PGC-1ß and mitochondrial gene expression are seen in vivo; fractionation of obese mouse adipose tissue reveals that PPARγ and PGC-1ß, but not PGC-1α, are coordinately up-regulated in adipocytes relative to preadipocytes and that TZD treatment induces PGC-1ß and mitochondrial marker genes in adipose tissue of obese mice. We propose that PPARγ directly induces PGC-1ß expression in adipocytes and that this effect regulates adipocyte mitochondrial activity.


Asunto(s)
Adipocitos/citología , PPAR gamma/metabolismo , Transactivadores/metabolismo , Células 3T3-L1 , Tejido Adiposo/metabolismo , Animales , Ácidos Grasos/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Obesos , Mitocondrias/metabolismo , Modelos Biológicos , Consumo de Oxígeno , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Tiazolidinedionas/farmacología , Factores de Transcripción
7.
Proc Natl Acad Sci U S A ; 106(47): 19854-9, 2009 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-19903885

RESUMEN

Identification of thyroid hormone receptor (TR) co-regulators has enhanced our understanding of thyroid hormone (TH) action. However, it is likely that many other co-regulators remained unidentified, and unbiased methods are required to discover these proteins. We have previously demonstrated that the yeast Saccharomyces cerevisiae is an excellent system in which to study TR action, and that defined TR signaling complexes in a eukaryotic background devoid of complicating influences of mammalian cell co-regulators can be constructed and analyzed for endogenous yeast genes, many of which are conserved in mammals. Here, a modified synthetic genetic array analysis was performed by crossing a yeast strain that expressed TRbeta1 and the co-activator GRIP1/SRC2 with 384 yeast strains bearing deletions of known genes. Eight genes essential for TH action were isolated, of which 4 are conserved in mammals. Examination of one, the yeast CCR4 and its human homolog CCR4/NOT6 (hCCR4), confirmed that (i) transfected CCR4 potentiates a TH response in cultured cells more efficiently than established TR co-activators and (ii) knockdown of CCR4 expression strongly inhibited a TH response (>80%). TH treatment promoted rapid and sustained hCCR4 recruitment to the TH-responsive deiodinase 1 promoter and TR co-localizes with hCCR4 in the nucleus and interacts with hCCR4 in 2-hybrid and pull-down assays. These findings indicate that a modified yeast synthetic genetic array strategy is a feasible method for unbiased identification of conserved genes essential for TR and other nuclear receptor hormone functions in mammals.


Asunto(s)
Análisis por Micromatrices/métodos , Receptores CCR4/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae , Receptores beta de Hormona Tiroidea/metabolismo , Animales , Regulación Fúngica de la Expresión Génica , Células HeLa , Humanos , Coactivador 2 del Receptor Nuclear/genética , Coactivador 2 del Receptor Nuclear/metabolismo , Regiones Promotoras Genéticas , Receptores CCR4/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Transducción de Señal/fisiología , Receptores beta de Hormona Tiroidea/genética , Triyodotironina/metabolismo
8.
Proc Natl Acad Sci U S A ; 106(49): 20717-22, 2009 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-19926848

RESUMEN

Nuclear receptors are important targets for pharmaceuticals, but similarities between family members cause difficulties in obtaining highly selective compounds. Synthetic ligands that are selective for thyroid hormone (TH) receptor beta (TRbeta) vs. TRalpha reduce cholesterol and fat without effects on heart rate; thus, it is important to understand TRbeta-selective binding. Binding of 3 selective ligands (GC-1, KB141, and GC-24) is characterized at the atomic level; preferential binding depends on a nonconserved residue (Asn-331beta) in the TRbeta ligand-binding cavity (LBC), and GC-24 gains extra selectivity from insertion of a bulky side group into an extension of the LBC that only opens up with this ligand. Here we report that the natural TH 3,5,3'-triodothyroacetic acid (Triac) exhibits a previously unrecognized mechanism of TRbeta selectivity. TR x-ray structures reveal better fit of ligand with the TRalpha LBC. The TRbeta LBC, however, expands relative to TRalpha in the presence of Triac (549 A(3) vs. 461 A(3)), and molecular dynamics simulations reveal that water occupies the extra space. Increased solvation compensates for weaker interactions of ligand with TRbeta and permits greater flexibility of the Triac carboxylate group in TRbeta than in TRalpha. We propose that this effect results in lower entropic restraint and decreases free energy of interactions between Triac and TRbeta, explaining subtype-selective binding. Similar effects could potentially be exploited in nuclear receptor drug design.


Asunto(s)
Entropía , Receptores alfa de Hormona Tiroidea/metabolismo , Receptores beta de Hormona Tiroidea/metabolismo , Ácido Acético/química , Ácido Acético/metabolismo , Sitios de Unión , Humanos , Enlace de Hidrógeno , Ligandos , Simulación de Dinámica Molecular , Docilidad , Electricidad Estática , Termodinámica , Triyodotironina/química , Triyodotironina/metabolismo , Agua
9.
Hepatology ; 52(6): 2001-11, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20938947

RESUMEN

UNLABELLED: Nonalcoholic fatty liver disease (NAFLD) is a common complication of obesity that can progress to nonalcoholic steatohepatitis (NASH), a serious liver pathology that can advance to cirrhosis. The mechanisms responsible for NAFLD progression to NASH remain unclear. Lack of a suitable animal model that faithfully recapitulates the pathophysiology of human NASH is a major obstacle in delineating mechanisms responsible for progression of NAFLD to NASH and, thus, development of better treatment strategies. We identified and characterized a novel mouse model, middle-aged male low-density lipoprotein receptor (LDLR)(-/-) mice fed a high-fat diet (HFD), which developed NASH associated with four of five metabolic syndrome (MS) components. In these mice, as observed in humans, liver steatosis and oxidative stress promoted NASH development. Aging exacerbated the HFD-induced NASH such that liver steatosis, inflammation, fibrosis, oxidative stress, and liver injury markers were greatly enhanced in middle-aged versus young LDLR(-/-) mice. Although expression of genes mediating fatty acid oxidation and antioxidant responses were up-regulated in young LDLR(-/-) mice fed HFD, they were drastically reduced in MS mice. However, similar to recent human trials, NASH was partially attenuated by an insulin-sensitizing peroxisome proliferator-activated receptor-gamma (PPARγ) ligand, rosiglitazone. In addition to expected improvements in MS, newly identified mechanisms of PPARγ ligand effects included stimulation of antioxidant gene expression and mitochondrial ß-oxidation, and suppression of inflammation and fibrosis. LDLR-deficiency promoted NASH, because middle-aged C57BL/6 mice fed HFD did not develop severe inflammation and fibrosis, despite increased steatosis. CONCLUSION: MS mice represent an ideal model to investigate NASH in the context of MS, as commonly occurs in human disease, and NASH development can be substantially attenuated by PPARγ activation, which enhances ß-oxidation.


Asunto(s)
Hígado Graso/prevención & control , Receptores de LDL/deficiencia , Tiazolidinedionas/uso terapéutico , Envejecimiento/fisiología , Animales , Antioxidantes/metabolismo , Grasas de la Dieta/efectos adversos , Hígado Graso/genética , Expresión Génica , Hepatitis/etiología , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Síndrome Metabólico , Ratones , Ratones Noqueados , Mitocondrias Hepáticas/fisiología , Estrés Oxidativo , PPAR gamma/metabolismo , Rosiglitazona
10.
Proc Natl Acad Sci U S A ; 105(2): 663-7, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18160532

RESUMEN

Atherosclerotic cardiovascular disease is a major problem despite the availability of drugs that influence major risk factors. New treatments are needed, and there is growing interest in therapies that may have multiple actions. Thyroid hormone modulates several cardiovascular risk factors and delays atherosclerosis progression in humans. However, use of thyroid hormone is limited by side effects, especially in the heart. To overcome this limitation, pharmacologically selective thyromimetics that mimic metabolic effects of thyroid hormone and bypass side effects are under development. In animal models, such thyromimetics have been shown to stimulate cholesterol elimination through LDL and HDL pathways and decrease body weight without eliciting side effects. We report here studies on a selective thyromimetic [KB2115; (3-[[3,5-dibromo-4-[4-hydroxy-3-(1-methylethyl)-phenoxy]-phenyl]-amino]-3-oxopropanoic acid)] in humans. In moderately overweight and hypercholesterolemic subjects KB2115 was found to be safe and well tolerated and elicited up to a 40% lowering of total and LDL cholesterol after 14 days of treatment. Bile acid synthesis was stimulated without evidence of increased cholesterol production, indicating that KB2115 induced net cholesterol excretion. KB2115 did not provoke detectable effects on the heart, suggesting that the pharmacological selectivity observed in animal models translates to humans. Thus, selective thyromimetics deserve further study as agents to treat dyslipidemia and other risk factors for atherosclerosis.


Asunto(s)
Anilidas/farmacología , Ácidos y Sales Biliares/metabolismo , LDL-Colesterol/metabolismo , Corazón/efectos de los fármacos , Glándula Tiroides/efectos de los fármacos , Hormonas Tiroideas/metabolismo , Adolescente , Adulto , Anilidas/química , Método Doble Ciego , Femenino , Humanos , Metabolismo de los Lípidos , Masculino , Persona de Mediana Edad , Placebos , Factores de Riesgo , Tiazolidinedionas/farmacología
11.
Proteins ; 75(1): 111-7, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18798561

RESUMEN

Protein quality and stability are critical during protein purification for X-ray crystallography. A target protein that is easy to manipulate and crystallize becomes a valuable product useful for high-throughput crystallography for drug design and discovery. In this work, a single surface mutation, D355R, was shown to be crucial for converting the modestly stable monomeric ligand binding domain of the human thyroid hormone receptor (TR LBD) into a stable dimer. The structure of D335R TR LBD mutant was solved using X-ray crystallography and refined to 2.2 A resolution with R(free)/R values of 24.5/21.7. The crystal asymmetric unit reveals the TR dimer with two molecules of the hormone-bound LBD related by twofold symmetry. The ionic interface between the two LBDs comprises residues within loop H10-H11 and loop H6-H7 as well as the C-terminal halves of helices 8 of both protomers. Direct intermolecular contacts formed between the introduced residue Arg 355 of one TR molecule and Glu 324 of the second molecule become a part of the extended dimerization interface of 1330 A(2) characteristic for a strong complex assembly that is additionally strengthened by buffer solutes.


Asunto(s)
Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Receptores de Hormona Tiroidea/genética , Receptores de Hormona Tiroidea/metabolismo , Cristalografía por Rayos X , Humanos , Ligandos , Proteínas Mutantes/química , Mutación Puntual , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Estabilidad Proteica , Receptores de Hormona Tiroidea/química
12.
Biochemistry ; 47(28): 7465-76, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18558711

RESUMEN

The potency and efficacy of ligands for nuclear receptors (NR) result both from the affinity of the ligand for the receptor and from the affinity that various coregulatory proteins have for ligand-receptor complexes; the latter interaction, however, is rarely quantified. To understand the molecular basis for ligand potency and efficacy, we developed dual time-resolved fluorescence resonance energy transfer (tr-FRET) assays and quantified binding of both ligand and coactivator or corepressor to the thyroid hormone receptor (TR). Promoter-bound TR exerts dual transcriptional regulatory functions, recruiting corepressor proteins and repressing transcription in the absence of thyroid hormones (THs) and shedding corepressors in favor of coactivators upon binding agonists, activating transcription. Our tr-FRET assays involve a TRE sequence labeled with terbium (fluorescence donor), TRbeta.RXRalpha heterodimer, and fluorescein-labeled NR interaction domains of coactivator SRC3 or corepressor NCoR (fluorescence acceptors). Through coregulator titrations, we could determine the affinity of SRC3 or NCoR for TRE-bound TR.RXR heterodimers, unliganded or saturated with different THs. Alternatively, through ligand titrations, we could determine the relative potencies of different THs. The order of TR agonist potencies is as follows: GC-1 approximately T 3 approximately TRIAC approximately T 4 >> rT 3 (for both coactivator recruitment and corepressor dissociation); the affinities of SRC3 binding to TR-ligand complexes followed a similar trend. This highlights the fact that the low activity of rT 3 is derived both from its low affinity for TR and from the low affinity of SRC for the TR-rT 3 complex. The TR antagonist NH-3 failed to induce SRC3 recruitment but did effect NCoR dissociation. These assays provide quantitative information about the affinity of two key interactions that are determinants of NR ligand potency and efficacy.


Asunto(s)
Histona Acetiltransferasas/metabolismo , Proteínas Nucleares/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Proteínas Represoras/metabolismo , Transactivadores/metabolismo , Secuencia de Bases , Sitios de Unión , Humanos , Cinética , Ligandos , Datos de Secuencia Molecular , Peso Molecular , Co-Represor 1 de Receptor Nuclear , Coactivador 3 de Receptor Nuclear , Regiones Promotoras Genéticas , Receptores de Hormona Tiroidea/química , Receptores de Hormona Tiroidea/genética , Espectrometría de Fluorescencia , Tiroxina/metabolismo , Triyodotironina/metabolismo
13.
BMC Struct Biol ; 8: 8, 2008 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-18237438

RESUMEN

BACKGROUND: Thyroid receptors, TRalpha and TRbeta, are involved in important physiological functions such as metabolism, cholesterol level and heart activities. Whereas metabolism increase and cholesterol level lowering could be achieved by TRbeta isoform activation, TRalpha activation affects heart rates. Therefore, beta-selective thyromimetics have been developed as promising drug-candidates for treatment of obesity and elevated cholesterol level. GC-1 [3,5-dimethyl-4-(4'-hydroxy-3'-isopropylbenzyl)-phenoxy acetic acid] has ability to lower LDL cholesterol with 600- to 1400-fold more potency and approximately two- to threefold more efficacy than atorvastatin (Lipitor(c)) in studies in rats, mice and monkeys. RESULTS: To investigate GC-1 specificity, we solved crystal structures and performed molecular dynamics simulations of both isoforms complexed with GC-1. Crystal structures reveal that, in TRalpha Arg228 is observed in multiple conformations, an effect triggered by the differences in the interactions between GC-1 and Ser277 or the corresponding asparagine (Asn331) of TRbeta. The corresponding Arg282 of TRbeta is observed in only one single stable conformation, interacting effectively with the ligand. Molecular dynamics support this model: our simulations show that the multiple conformations can be observed for the Arg228 in TRalpha, in which the ligand interacts either strongly with the ligand or with the Ser277 residue. In contrast, a single stable Arg282 conformation is observed for TRbeta, in which it strongly interacts with both GC-1 and the Asn331. CONCLUSION: Our analysis suggests that the key factors for GC-1 selectivity are the presence of an oxyacetic acid ester oxygen and the absence of the amino group relative to T3. These results shed light into the beta-selectivity of GC-1 and may assist the development of new compounds with potential as drug candidates to the treatment of hypercholesterolemia and obesity.


Asunto(s)
Acetatos/química , Fenoles/química , Receptores alfa de Hormona Tiroidea/química , Receptores beta de Hormona Tiroidea/química , Acetatos/metabolismo , Sitios de Unión , Simulación por Computador , Cristalografía por Rayos X , Células HeLa , Humanos , Ligandos , Modelos Biológicos , Fenoles/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Receptores alfa de Hormona Tiroidea/metabolismo , Receptores beta de Hormona Tiroidea/metabolismo
14.
Mol Cell Endocrinol ; 296(1-2): 69-77, 2008 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-18930112

RESUMEN

It is desirable to obtain new antagonists for thyroid hormone receptors (TRs) and other nuclear receptors (NRs). We previously used X-ray structural models of TR ligand binding domains (LBDs) to design compounds, such as NH-3, that impair coactivator binding to activation function 2 (AF-2) and block thyroid hormone (triiodothyronine, T(3)) actions. However, TRs bind DNA and are transcriptionally active without ligand. Thus, NH-3 could modulate TR activity via effects on other coregulator interaction surfaces, such as activation function (AF-1) and corepressor binding sites. Here, we find that NH-3 blocks TR-LBD interactions with coactivators and corepressors and also inhibits activities of AF-1 and AF-2 in transfections. While NH-3 lacks detectable agonist activity at T(3)-activated genes in GC pituitary cells it nevertheless activates spot 14 (S14) in HTC liver cells with the latter effect accompanied by enhanced histone H4 acetylation and coactivator recruitment at the S14 promoter. Surprisingly, T(3) promotes corepressor recruitment to target promoters. NH-3 effects vary; we observe transient recruitment of N-CoR to S14 in GC cells and dismissal and rebinding of N-CoR to the same promoter in HTC cells. We propose that NH-3 will generally behave as an antagonist by blocking AF-1 and AF-2 but that complex effects on coregulator recruitment may result in partial/mixed agonist effects that are independent of blockade of T(3) binding in some contexts. These properties could ultimately be utilized in drug design and development of new selective TR modulators.


Asunto(s)
Acetatos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de los fármacos , Receptores de Hormona Tiroidea/antagonistas & inhibidores , Acetilación/efectos de los fármacos , Línea Celular , Histonas/metabolismo , Humanos , Modelos Biológicos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiología , Co-Represor 1 de Receptor Nuclear , Fenoxiacetatos , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína/fisiología , Receptores de Hormona Tiroidea/química , Receptores de Hormona Tiroidea/metabolismo , Proteínas Represoras/metabolismo , Proteínas Represoras/fisiología , Transactivadores/metabolismo , Transactivadores/fisiología , Factores de Transcripción/genética , Transfección
15.
J Steroid Biochem Mol Biol ; 111(3-5): 262-7, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18621127

RESUMEN

Selective thyroid hormone receptor subtype-beta (TRbeta) agonists have received attention as potential treatments for hypercholesterolemia and obesity, but have received less attention as treatments for diabetes, partly because this condition is not improved in thyroid hormone excess states. The TRbeta selective agonist KB-141 induces 5-10% increases in metabolic rate and lowering of plasma cholesterol levels without tachycardia in lean rats, unlike the major active thyroid hormone, T3. In the current study, we determined whether KB-141 promotes weight loss in obese animals and whether it exhibits anti-diabetogenic effects. Body weight, adiposity (DEXA), and lipid levels were examined following p.o. administration of KB-141 to obese Zucker fa/fa rats at 0.00547-0.547 mg/kg/day for 21 days, and in ob/ob mice at 0.5mg/kg/day KB-141 for 7 days. In rats, KB-141 reduced body weight by 6 and 8%, respectively, at 0.167 and 0.0547 mg/kg/day without tachycardia and adiposity was reduced at 0.167 mg/kg/day (5-6%). In ob/ob mice, KB-141 lowered serum cholesterol (35%), triacylglycerols (35%) and both serum and hepatic free fatty acids (18-20%) without tachycardia. Treatment of ob/ob mice with KB-141 (0.0547 or 0.328 mg/kg/day over 2 weeks) improved glucose tolerance and insulin sensitivity in a dose-dependent manner with no effect on heart rate. Thus, KB-141 elicits anti-obesity, lipid lowering and anti-diabetic effects without tachycardia suggesting that selective TRbeta activation may be useful strategy to attenuate features of the metabolic syndrome.


Asunto(s)
Fármacos Antiobesidad/uso terapéutico , Diabetes Mellitus/tratamiento farmacológico , Hipercolesterolemia/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Hipolipemiantes/uso terapéutico , Obesidad/tratamiento farmacológico , Éteres Fenílicos/uso terapéutico , Fenilacetatos/uso terapéutico , Receptores beta de Hormona Tiroidea/agonistas , Animales , Fármacos Antiobesidad/química , Fármacos Antiobesidad/farmacología , Peso Corporal/efectos de los fármacos , Femenino , Humanos , Hipoglucemiantes/química , Hipoglucemiantes/farmacología , Hipolipemiantes/química , Hipolipemiantes/farmacología , Ratones , Ratones Obesos , Estructura Molecular , Éteres Fenílicos/química , Éteres Fenílicos/farmacología , Fenilacetatos/química , Fenilacetatos/farmacología , Ratas , Ratas Zucker
16.
Mol Endocrinol ; 21(12): 2919-28, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17823305

RESUMEN

The development of nuclear hormone receptor antagonists that directly inhibit the association of the receptor with its essential coactivators would allow useful manipulation of nuclear hormone receptor signaling. We previously identified 3-(dibutylamino)-1-(4-hexylphenyl)-propan-1-one (DHPPA), an aromatic beta-amino ketone that inhibits coactivator recruitment to thyroid hormone receptor beta (TRbeta), in a high-throughput screen. Initial evidence suggested that the aromatic beta-enone 1-(4-hexylphenyl)-prop-2-en-1-one (HPPE), which alkylates a specific cysteine residue on the TRbeta surface, is liberated from DHPPA. Nevertheless, aspects of the mechanism and specificity of action of DHPPA remained unclear. Here, we report an x-ray structure of TRbeta with the inhibitor HPPE at 2.3-A resolution. Unreacted HPPE is located at the interface that normally mediates binding between TRbeta and its coactivator. Several lines of evidence, including experiments with TRbeta mutants and mass spectroscopic analysis, showed that HPPE specifically alkylates cysteine residue 298 of TRbeta, which is located near the activation function-2 pocket. We propose that this covalent adduct formation proceeds through a two-step mechanism: 1) beta-elimination to form HPPE; and 2) a covalent bond slowly forms between HPPE and TRbeta. DHPPA represents a novel class of potent TRbeta antagonist, and its crystal structure suggests new ways to design antagonists that target the assembly of nuclear hormone receptor gene-regulatory complexes and block transcription.


Asunto(s)
Receptores de Hormona Tiroidea/antagonistas & inhibidores , Receptores de Hormona Tiroidea/metabolismo , Acetona/química , Acetona/farmacología , Secuencia de Aminoácidos , Sitios de Unión , Cisteína/genética , Cisteína/metabolismo , Humanos , Cetonas/química , Ligandos , Espectrometría de Masas , Modelos Moleculares , Datos de Secuencia Molecular , Mutación/genética , Unión Proteica , Estructura Terciaria de Proteína
17.
N Engl J Med ; 349(9): 837-46, 2003 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-12944569

RESUMEN

BACKGROUND: We compared two strategies for treating patients infected with multidrug-resistant human immunodeficiency virus (HIV). METHODS: Patients with multidrug-resistant HIV and HIV RNA levels of more than 5000 copies per milliliter were randomly assigned to a four-month structured interruption of treatment followed by a change in antiretroviral regimen (treatment-interruption group) or to an immediate change in regimen (control group). Genotypic and phenotypic resistance testing was performed. Disease progression, death, and changes in genotypic resistance, CD4 cell counts, HIV RNA levels, and quality of life were assessed. RESULTS: After a median follow-up of 11.6 months, disease progression or death occurred in 22 of the 138 patients in the treatment-interruption group and in 12 of the 132 patients in the control group (P=0.01), with a hazard ratio of 2.57 (95 percent confidence interval, 1.2 to 5.5) for the treatment-interruption group. There were eight deaths in each group. In the treatment-interruption group, the mutant HIV populations completely or partially reverted to wild type by four months in 64.0 percent of patients. As compared with the control group, the treatment-interruption group had a mean CD4 cell count that was 85 cells per cubic millimeter lower from months 0 through 4 (P<0.001), 47 cells per cubic millimeter lower from months 5 through 8 (P<0.001), and 31 cells per cubic millimeter lower after eight months (P=0.11). The mean HIV RNA levels were 1.2 log copies per milliliter higher (on a base-10 scale) in the treatment-interruption group during months 0 through 4 (P<0.001), but they were not significantly different from those in the control group after month 4. The overall quality of life was similar in the two groups. CONCLUSIONS: In patients infected with multidrug-resistant HIV, structured interruption of treatment was associated with greater progression of disease and did not confer immunologic or virologic benefits or improve the overall quality of life.


Asunto(s)
Fármacos Anti-VIH/administración & dosificación , Infecciones por VIH/tratamiento farmacológico , Infecciones Oportunistas Relacionadas con el SIDA/epidemiología , Adulto , Recuento de Linfocito CD4 , Progresión de la Enfermedad , Esquema de Medicación , Farmacorresistencia Viral Múltiple , Femenino , VIH/efectos de los fármacos , VIH/genética , VIH/aislamiento & purificación , Infecciones por VIH/mortalidad , Humanos , Masculino , Persona de Mediana Edad , Análisis Multivariante , ARN Viral/sangre
18.
J Mol Biol ; 360(3): 586-98, 2006 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-16781732

RESUMEN

The thyroid hormone receptor (TR) D-domain links the ligand-binding domain (LBD, EF-domain) to the DNA-binding domain (DBD, C-domain), but its structure, and even its existence as a functional unit, are controversial. The D domain is poorly conserved throughout the nuclear receptor family and was originally proposed to comprise an unfolded hinge that facilitates rotation between the LBD and the DBD. Previous TR LBD structures, however, have indicated that the true unstructured region is three to six amino acid residues long and that the D-domain N terminus folds into a short amphipathic alpha-helix (H0) contiguous with the DBD and that the C terminus of the D-domain comprises H1 and H2 of the LBD. Here, we solve structures of TR-LBDs in different crystal forms and show that the N terminus of the TRalpha D-domain can adopt two structures; it can either fold into an amphipathic helix that resembles TRbeta H0 or form an unstructured loop. H0 formation requires contacts with the AF-2 coactivator-binding groove of the neighboring TR LBD, which binds H0 sequences that resemble coactivator LXXLL motifs. Structural analysis of a liganded TR LBD with small angle X-ray scattering (SAXS) suggests that AF-2/H0 interactions mediate dimerization of this protein in solution. We propose that the TR D-domain has the potential to form functionally important extensions of the DBD and LBD or unfold to permit TRs to adapt to different DNA response elements. We also show that mutations of the D domain LXXLL-like motif indeed selectively inhibit TR interactions with an inverted palindromic response element (F2) in vitro and TR activity at this response element in cell-based transfection experiments.


Asunto(s)
Receptores alfa de Hormona Tiroidea/química , Receptores alfa de Hormona Tiroidea/metabolismo , Receptores beta de Hormona Tiroidea/química , Receptores beta de Hormona Tiroidea/metabolismo , Secuencias de Aminoácidos , ADN/metabolismo , Dimerización , Células HeLa , Humanos , Ligandos , Modelos Moleculares , Unión Proteica , Pliegue de Proteína , Estructura Terciaria de Proteína , Elementos de Respuesta/genética , Soluciones , Relación Estructura-Actividad , Triyodotironina/metabolismo , Células Tumorales Cultivadas , Difracción de Rayos X
19.
Sci STKE ; 2006(341): pl3, 2006 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-16804159

RESUMEN

To provide alternative methods for regulation of gene transcription initiated by the binding of thyroid hormone (T3) to the thyroid receptor (TR), we have developed a high-throughput method for discovering inhibitors of the interaction of TR with its transcriptional coactivators. The screening method is based on fluorescence polarization (FP), one of the most sensitive and robust high-throughput methods for the study of protein-protein interactions. A fluorescently labeled coactivator is excited by polarized light. The emitted polarized light is a function of the molecular properties of the labeled coactivator, especially Brownian molecular rotation, which is very sensitive to changes in the molecular mass of the labeled complex. Dissociation of hormone receptor from fluorescently labeled coactivator peptide in the presence of small molecules can be detected by this competition method, and the assay can be performed in a high-throughput screening format. Hit compounds identified by this method are evaluated by several secondary assay methods, including a dose-response analysis, a semiquantitative glutathione-S-transferase assay, and a hormone displacement assay. Subsequent in vitro transcription assays can detect inhibition of thyroid signaling at low micromolar concentrations of small molecules in the presence of T3.


Asunto(s)
Acetiltransferasas/antagonistas & inhibidores , Evaluación Preclínica de Medicamentos/métodos , Polarización de Fluorescencia/métodos , Receptores de Hormona Tiroidea/efectos de los fármacos , Transactivadores/antagonistas & inhibidores , Acetiltransferasas/metabolismo , Unión Competitiva , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Glutatión Transferasa/análisis , Histona Acetiltransferasas , Humanos , Indicadores y Reactivos , Peso Molecular , Coactivador 3 de Receptor Nuclear , Concentración Osmolar , Fotoquímica , Unión Proteica , Receptores de Hormona Tiroidea/metabolismo , Rotación , Sensibilidad y Especificidad , Transactivadores/metabolismo
20.
HIV Clin Trials ; 8(6): 357-70, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18042501

RESUMEN

BACKGROUND: Treatment-naïve participants were randomized to three antiretroviral strategies (all with nucleoside reverse transcriptase inhibitor [NRTI] background): protease inhibitor (PI), non-nucleoside reverse transcriptase inhibitor (NNRTI), or PI+NNRTI. The strategies were compared for drug resistance at first virologic failure (VF; HIV RNA >1000 copies/mL). The impact of resistance on AIDS or death was determined. METHOD: Drug resistance was determined by genotype. Cox models were used to compare the strategies for VF with resistance and to determine the impact of resistance on AIDS or death. RESULTS: Of 1,360 participants, 866 experienced VF; 226 experienced AIDS or death (median follow-up 5 years). Rates (per 100 personyears) for VF with resistance were 14.9 (PI), 10.8 (NNRTI), and 11.5 (PI+NNRTI); hazard ratio (HR) was 0.78 (95% CI 0.61-0.99) for NNRTI versus PI. Compared to those with no VF, there was a significantly increased risk of AIDS or death for participants with solitary NNRTI resistance (HR 2.31, 95% CI 1.46-3.66) and for those failing with no known resistance (HR 1.78, 95% CI 1.18-2.68). Participants failing with solitary NNRTI resistance and with no resistance had the lowest percent of time on antiretroviral treatment (ART) and the lowest cumulative mean adherence scores. CONCLUSION: For treatment-naïve participants, the risk of AIDS or death is increased for those who failed virologically with solitary NNRTI resistance and those who failed with no known drug resistance compared to those with no virologic failure. Both the lack of ART exposure in nonadherent participants and the development of NNRTI resistance among those who take and fail their ART regimen predict poor clinical outcomes.


Asunto(s)
Fármacos Anti-VIH/uso terapéutico , Farmacorresistencia Viral/genética , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/virología , VIH/genética , Terapia Antirretroviral Altamente Activa , Progresión de la Enfermedad , Infecciones por VIH/mortalidad , Humanos , Incidencia , Cooperación del Paciente , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA