Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Invest New Drugs ; 40(5): 1001-1010, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35802288

RESUMEN

Preclinical models suggest anticancer activity of IM156, a novel biguanide mitochondrial protein complex 1 inhibitor of oxidative phosphorylation (OXPHOS). This first-in-human dose-escalation study enrolled patients with refractory advanced solid tumors to determine the maximum tolerated dose (MTD) or recommended phase 2 dose (RP2D). Eligible patients received oral IM156 every other day (QOD) or daily (QD) and were assessed for safety, dose-limiting toxicities (DLTs), pharmacokinetics, and preliminary signals of efficacy. 22 patients with advanced cancers (gastric, n = 8; colorectal, n = 3; ovarian, n = 3; other, n = 8) received IM156 100 to 1,200 mg either QOD or QD. There were no DLTs. However, 1,200 mg QD was not well tolerated due to nausea; 800 mg QD was determined as the RP2D. The most frequent treatment-related AEs (TRAEs) were nausea (n = 15; 68%), diarrhea (n = 10; 46%), emesis (n = 9; 41%), fatigue (n = 4; 18%) and abdominal pain, constipation, and blood lactate increased (n = 2 each; 9%). Grade 3 nausea (n = 3; 14%) was the only grade ≥ 3 TRAE. Plasma exposures increased dose proportionally; mean Day 27 area under the curve (AUC0-24) values were higher following QD administration compared to the respective QOD regimen. Stable disease (SD), observed in 7 (32%) patients (confirmed in 2 [9%]), was the best response. To our knowledge, this is the first phase 1 study of an OXPHOS inhibitor that established a RP2D for further clinical development in cancer. Observed AEs of IM156 were manageable and SD was the best response.


Asunto(s)
Antineoplásicos , Neoplasias , Antineoplásicos/efectos adversos , Biguanidas/uso terapéutico , Relación Dosis-Respuesta a Droga , Humanos , Dosis Máxima Tolerada , Náusea/inducido químicamente , Neoplasias/metabolismo , Fosforilación Oxidativa
2.
J Pharmacol Exp Ther ; 379(3): 290-300, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34593558

RESUMEN

Metabolic reprogramming of the myofibroblast plays a fundamental role in the pathogenesis of fibrosing interstitial lung diseases. Here, we characterized the in vitro and in vivo metabolic and antifibrotic effects of IM156, an oxidative phosphorylation (OXPHOS) modulator that acts by inhibiting protein complex 1. In vitro, IM156 inhibited transforming growth factor ß (TGFß)-dependent increases in mitochondrial oxygen consumption rate and expression of myofibroblast markers in human pulmonary fibroblasts without altering cell viability or adding to TGFß-induced increases in the extracellular acidification rate. IM156 significantly increased cellular AMP-activated protein kinase (AMPK) phosphorylation and was 60-fold more potent than metformin. In vivo, chronic oral administration of IM156 was highly distributed to major peripheral organs (i.e., lung, liver, kidney, heart) and had significant dose-related effects on the plasma metabolome consistent with OXPHOS modulation and AMPK activation. IM156 increased glycolysis, lipolysis, ß-oxidation, and amino acids and decreased free fatty acids, tricarboxylic acid cycle activity, and protein synthesis. In the murine bleomycin model of pulmonary fibrosis, daily oral administration of IM156, administered 7 days after lung injury, attenuated body/lung weight changes and reduced lung fibrosis and inflammatory cell infiltration. The plasma exposures of IM156 were comparable to well tolerated doses in human studies. In conclusion, the metabolic and antifibrotic effects of IM156 suggest that OXPHOS modulation can attenuate myofibroblast metabolic reprogramming and support testing IM156 as a therapy for idiopathic pulmonary fibrosis and other fibrotic diseases. SIGNIFICANCE STATEMENT: Fibrosing interstitial lung diseases have a poor prognosis, and current antifibrotic treatments have significant limitations. This study demonstrates that attenuation of fibrogenic metabolic remodeling, by modulation of oxidative phosphorylation with IM156, prevents myofibroblast phenotype/collagen deposition and is a potentially effective and translational antifibrotic strategy.


Asunto(s)
Antifibróticos/farmacología , Reprogramación Celular/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Fibrosis Pulmonar/metabolismo , Animales , Antifibróticos/química , Antifibróticos/uso terapéutico , Línea Celular , Reprogramación Celular/fisiología , Relación Dosis-Respuesta a Droga , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Masculino , Metabolómica/métodos , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/prevención & control
3.
Cancer Discov ; 8(9): 1096-1111, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29903880

RESUMEN

Clinically relevant subtypes exist for pancreatic ductal adenocarcinoma (PDAC), but molecular characterization is not yet standard in clinical care. We implemented a biopsy protocol to perform time-sensitive whole-exome sequencing and RNA sequencing for patients with advanced PDAC. Therapeutically relevant genomic alterations were identified in 48% (34/71) and pathogenic/likely pathogenic germline alterations in 18% (13/71) of patients. Overall, 30% (21/71) of enrolled patients experienced a change in clinical management as a result of genomic data. Twenty-six patients had germline and/or somatic alterations in DNA-damage repair genes, and 5 additional patients had mutational signatures of homologous recombination deficiency but no identified causal genomic alteration. Two patients had oncogenic in-frame BRAF deletions, and we report the first clinical evidence that this alteration confers sensitivity to MAPK pathway inhibition. Moreover, we identified tumor/stroma gene expression signatures with clinical relevance. Collectively, these data demonstrate the feasibility and value of real-time genomic characterization of advanced PDAC.Significance: Molecular analyses of metastatic PDAC tumors are challenging due to the heterogeneous cellular composition of biopsy specimens and rapid progression of the disease. Using an integrated multidisciplinary biopsy program, we demonstrate that real-time genomic characterization of advanced PDAC can identify clinically relevant alterations that inform management of this difficult disease. Cancer Discov; 8(9); 1096-111. ©2018 AACR.See related commentary by Collisson, p. 1062This article is highlighted in the In This Issue feature, p. 1047.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Perfilación de la Expresión Génica/métodos , Variación Genética , Genómica/métodos , Neoplasias Pancreáticas/genética , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Ductal Pancreático/tratamiento farmacológico , Reparación del ADN , Femenino , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Mutación de Línea Germinal , Recombinación Homóloga , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/tratamiento farmacológico , Medicina de Precisión , Análisis de Secuencia de ARN/métodos , Secuenciación del Exoma/métodos
4.
J Clin Invest ; 128(7): 2927-2943, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29863500

RESUMEN

Although nonmalignant stromal cells facilitate tumor growth and can occupy up to 90% of a solid tumor mass, better strategies to exploit these cells for improved cancer therapy are needed. Here, we describe a potent MMAE-linked antibody-drug conjugate (ADC) targeting tumor endothelial marker 8 (TEM8, also known as ANTXR1), a highly conserved transmembrane receptor broadly overexpressed on cancer-associated fibroblasts, endothelium, and pericytes. Anti-TEM8 ADC elicited potent anticancer activity through an unexpected killing mechanism we term DAaRTS (drug activation and release through stroma), whereby the tumor microenvironment localizes active drug at the tumor site. Following capture of ADC prodrug from the circulation, tumor-associated stromal cells release active MMAE free drug, killing nearby proliferating tumor cells in a target-independent manner. In preclinical studies, ADC treatment was well tolerated and induced regression and often eradication of multiple solid tumor types, blocked metastatic growth, and prolonged overall survival. By exploiting TEM8+ tumor stroma for targeted drug activation, these studies reveal a drug delivery strategy with potential to augment therapies against multiple cancer types.


Asunto(s)
Inmunoconjugados/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Receptores de Superficie Celular/antagonistas & inhibidores , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/deficiencia , Biomarcadores de Tumor/genética , Brentuximab Vedotina , Línea Celular Tumoral , Femenino , Humanos , Inmunoconjugados/farmacocinética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Ratones SCID , Proteínas de Microfilamentos , Neoplasias/metabolismo , Receptores de Péptidos/antagonistas & inhibidores , Receptores de Péptidos/deficiencia , Receptores de Péptidos/genética , Células del Estroma/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cancer Chemother Pharmacol ; 81(6): 1129-1141, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29603015

RESUMEN

PURPOSE: The aim of this analysis was to investigate the potential for ulixertinib (BVD-523) to prolong cardiac repolarization. The mean prolongation of the corrected QT (QTc) interval was predicted at the mean maximum drug concentrations of the recommended phase 2 dose (RP2D; 600 mg BID) and of higher concentrations. In addition, the effect of ulixertinib on other quantitative ECG parameters was assessed. METHODS: In a two-part, phase 1, open-label study in adults with advanced solid tumors, 105 patients [24 in Part 1 (dose escalation) and 81 in Part 2 (cohort expansion)] were included in a QT prolongation analysis. Electrocardiograms (ECGs) extracted from 12-lead Holter monitors, along with time-matched pharmacokinetic blood samples, were collected over 12 h on cycle 1 day 1 and cycle 1 day 15 and analyzed by a core ECG laboratory. RESULTS: A small increase in heart rate was observed on both study days (up to 5.6 bpm on day 1 and up to 7 bpm on day 15). The estimated mean changes from baseline in the study-specific QTc interval (QTcSS), at the ulixertinib Cmax, were - 0.529 ms (90% CI - 6.621, 5.562) on day 1 and - 9.202 ms (90% CI - 22.505, 4.101) on day 15. The concentration: QTc regression slopes were mildly positive but not statistically significant [0.53 (90% CI - 1.343, 2.412) and 1.16 (90% CI - 1.732, 4.042) ms per µg/mL for days 1 and 15, respectively]. Ulixertinib had no meaningful effect on PR or QRS intervals. CONCLUSIONS: Ulixertinib administered to patients with solid tumors at clinically relevant doses has a low risk for QT/QTc prolongation or any other effects on ECG parameters. REGISTRATION: The study is registered at Clinicaltrials.gov (NCT01781429) and was sponsored by BioMed Valley Discoveries.


Asunto(s)
Aminopiridinas/administración & dosificación , Frecuencia Cardíaca/efectos de los fármacos , Síndrome de QT Prolongado/etiología , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/administración & dosificación , Pirroles/administración & dosificación , Anciano , Aminopiridinas/efectos adversos , Relación Dosis-Respuesta a Droga , Electrocardiografía Ambulatoria , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/efectos adversos , Pirroles/efectos adversos
6.
Cancer Discov ; 8(2): 184-195, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29247021

RESUMEN

Ulixertinib (BVD-523) is an ERK1/2 kinase inhibitor with potent preclinical activity in BRAF- and RAS-mutant cell lines. In this multicenter phase I trial (NCT01781429), 135 patients were enrolled to an accelerated 3 + 3 dose-escalation cohort and six distinct dose-expansion cohorts. Dose escalation included 27 patients, dosed from 10 to 900 mg twice daily and established the recommended phase II dose (RP2D) of 600 mg twice daily. Ulixertinib exposure was dose proportional to the RP2D, which provided near-complete inhibition of ERK activity in whole blood. In the 108-patient expansion cohort, 32% of patients required dose reduction. The most common treatment-related adverse events were diarrhea (48%), fatigue (42%), nausea (41%), and dermatitis acneiform (31%). Partial responses were seen in 3 of 18 (17%) patients dosed at or above maximum tolerated dose and in 11 of 81 (14%) evaluable patients in dose expansion. Responses occurred in patients with NRAS-, BRAF V600-, and non-V600 BRAF-mutant solid tumors.Significance: Here, we describe the first-in-human dose-escalation study of an ERK1/2 inhibitor for the treatment of patients with advanced solid tumors. Ulixertinib has an acceptable safety profile with favorable pharmacokinetics and has shown early evidence of clinical activity in NRAS- and BRAF V600- and non-V600-mutant solid-tumor malignancies. Cancer Discov; 8(2); 184-95. ©2017 AACR.See related commentary by Smalley and Smalley, p. 140This article is highlighted in the In This Issue feature, p. 127.


Asunto(s)
Aminopiridinas/uso terapéutico , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/genética , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirroles/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Aminopiridinas/farmacología , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Estadificación de Neoplasias , Neoplasias/diagnóstico , Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Pirroles/farmacología , Tomografía Computarizada por Rayos X , Resultado del Tratamiento , Adulto Joven
7.
Mol Cancer Ther ; 16(11): 2351-2363, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28939558

RESUMEN

Aberrant activation of signaling through the RAS-RAF-MEK-ERK (MAPK) pathway is implicated in numerous cancers, making it an attractive therapeutic target. Although BRAF and MEK-targeted combination therapy has demonstrated significant benefit beyond single-agent options, the majority of patients develop resistance and disease progression after approximately 12 months. Reactivation of ERK signaling is a common driver of resistance in this setting. Here we report the discovery of BVD-523 (ulixertinib), a novel, reversible, ATP-competitive ERK1/2 inhibitor with high potency and ERK1/2 selectivity. In vitro BVD-523 treatment resulted in reduced proliferation and enhanced caspase activity in sensitive cells. Interestingly, BVD-523 inhibited phosphorylation of target substrates despite increased phosphorylation of ERK1/2. In in vivo xenograft studies, BVD-523 showed dose-dependent growth inhibition and tumor regression. BVD-523 yielded synergistic antiproliferative effects in a BRAFV600E-mutant melanoma cell line xenograft model when used in combination with BRAF inhibition. Antitumor activity was also demonstrated in in vitro and in vivo models of acquired resistance to single-agent and combination BRAF/MEK-targeted therapy. On the basis of these promising results, these studies demonstrate BVD-523 holds promise as a treatment for ERK-dependent cancers, including those whose tumors have acquired resistance to other treatments targeting upstream nodes of the MAPK pathway. Assessment of BVD-523 in clinical trials is underway (NCT01781429, NCT02296242, and NCT02608229). Mol Cancer Ther; 16(11); 2351-63. ©2017 AACR.


Asunto(s)
Aminopiridinas/administración & dosificación , Melanoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Proto-Oncogénicas B-raf/genética , Pirroles/administración & dosificación , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Melanoma/genética , Melanoma/patología , Ratones , Mutación , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cancer Cell ; 31(4): 501-515.e8, 2017 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-28399408

RESUMEN

Targeting the tumor vasculature with antibody-drug conjugates (ADCs) is a promising anti-cancer strategy that in order to be realized must overcome several obstacles, including identification of suitable targets and optimal warheads. Here, we demonstrate that the cell-surface protein CD276/B7-H3 is broadly overexpressed by multiple tumor types on both cancer cells and tumor-infiltrating blood vessels, making it a potentially ideal dual-compartment therapeutic target. In preclinical studies CD276 ADCs armed with a conventional MMAE warhead destroyed CD276-positive cancer cells, but were ineffective against tumor vasculature. In contrast, pyrrolobenzodiazepine-conjugated CD276 ADCs killed both cancer cells and tumor vasculature, eradicating large established tumors and metastases, and improving long-term overall survival. CD276-targeted dual-compartment ablation could aid in the development of highly selective broad-acting anti-cancer therapies.


Asunto(s)
Antígenos B7/genética , Antígenos B7/metabolismo , Inmunoconjugados/farmacología , Neoplasias/irrigación sanguínea , Animales , Antineoplásicos/inmunología , Antineoplásicos/farmacología , Antígenos B7/inmunología , Benzodiazepinas/farmacología , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Línea Celular Tumoral , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Femenino , Humanos , Inmunoconjugados/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Terapia Molecular Dirigida/métodos , Neoplasias/patología , Neoplasias/terapia , Oligopéptidos/farmacología , Pirroles/farmacología , Conejos
9.
World J Microbiol Biotechnol ; 30(11): 3003-10, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25085723

RESUMEN

The nucleoside analogue, 1-(2-deoxy-2-fluoro-1-D-arabinofuranosyl)-5-iodouracil (FIAU) is a substrate for thymidine kinase (TK), which is commonly expressed in bacteria. It is currently being investigated in clinical studies as an in vivo bacterial infection detection agent. In developing countries where imaging facilities are not readily available, deploying such technology can be a big hurdle. However, a portable ex vivo system might provide a good alternative. In an in vitro system, [(125)I]-FIAU incubated with bacteria is phosphorylated by TK, and is trapped within the bacteria, which can be detected by radioscintography. The suitability of this agent to be utilized as part of an ex vivo bacterial detection system was evaluated. In the first part of this report, the optimization of the incubation and detection condition using E. coli as a test case is described. Samples were incubated in a growth promoting medium containing the label, then after filtering and washing, the amount of radioactivity trapped on the filter was quantitated by a scintillation counter. As a proof of concept demonstration, blinded urine samples from urinary tract infection (UTI) patients and normal donors were tested in the FIAU system. Of the 13 UTI positive and 15 normal urine samples tested, there were 2 false negatives and 1 false positive, respectively. Potential explanations for the false positive and negatives as well as the commercialization possibility of this system will be discussed.


Asunto(s)
Arabinofuranosil Uracilo/análogos & derivados , Bacterias/aislamiento & purificación , Infecciones Bacterianas/diagnóstico , Técnicas Bacteriológicas/métodos , Sistemas de Atención de Punto , Radiometría/métodos , Arabinofuranosil Uracilo/metabolismo , Bacterias/metabolismo , Infecciones Bacterianas/microbiología , Errores Diagnósticos , Humanos , Infecciones Urinarias/diagnóstico , Orina/microbiología
10.
Drug Discov Today Ther Strateg ; 8(3-4): 77-83, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22768020

RESUMEN

The Pharmaceutical Assets Portal aims to facilitate industry-academic collaborations for discovery of new indications for compounds no longer being developed by pharmaceutical companies, through eliminating barriers to access such compounds. The Portal's enabling infrastructure includes a national investigator database; a Foci-of-Expertise browser; a material transfer agreement template; and a funding partner. Whereas the goal of creating a shared compound repository remains to be achieved, the Portal has established a mechanism to facilitate future drug repositioning opportunities.

11.
J Med Chem ; 53(18): 6653-80, 2010 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-20726512

RESUMEN

α-Sulfone-α-piperidine and α-tetrahydropyranyl hydroxamates were explored that are potent inhibitors of MMP's-2, -9, and -13 that spare MMP-1, with oral efficacy in inhibiting tumor growth in mice and left-ventricular hypertrophy in rats and in the bovine cartilage degradation ex vivo explant system. α-Piperidine 19v (SC-78080/SD-2590) was selected for development toward the initial indication of cancer, while α-piperidine and α-tetrahydropyranyl hydroxamates 19w (SC-77964) and 9i (SC-77774), respectively, were identified as backup compounds.


Asunto(s)
Antiinflamatorios/síntesis química , Antineoplásicos/síntesis química , Fármacos Cardiovasculares/síntesis química , Ácidos Hidroxámicos/síntesis química , Inhibidores de la Metaloproteinasa de la Matriz , Piperidinas/síntesis química , Piranos/síntesis química , Sulfonas/síntesis química , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Disponibilidad Biológica , Fármacos Cardiovasculares/química , Fármacos Cardiovasculares/farmacología , Cartílago Articular/efectos de los fármacos , Cartílago Articular/patología , Bovinos , Cristalografía por Rayos X , Humanos , Ácidos Hidroxámicos/química , Ácidos Hidroxámicos/farmacología , Hipertrofia Ventricular Izquierda/tratamiento farmacológico , Macaca fascicularis , Ratones , Ratones Desnudos , Modelos Moleculares , Estructura Molecular , Piperidinas/química , Piperidinas/farmacología , Piranos/química , Piranos/farmacología , Ratas , Relación Estructura-Actividad , Sulfonas/química , Sulfonas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
ACS Med Chem Lett ; 1(2): 59-63, 2010 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-24900177

RESUMEN

Hematopoietic prostaglandin D synthase (HPGDS) is primarly expressed in mast cells, antigen-presenting cells, and Th-2 cells. HPGDS converts PGH2 into PGD2, a mediator thought to play a pivotal role in airway allergy and inflammatory processes. In this letter, we report the discovery of an orally potent and selective inhibitor of HPGDS that reduces the antigen-induced response in allergic sheep.

13.
Mol Endocrinol ; 19(6): 1460-73, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15761029

RESUMEN

We investigated the coregulator (coactivator and corepressor) interactions with the mineralocorticoid receptor (MR) that lead to activation and inhibition of the receptor in the presence of agonist and/or antagonist. Our results indicate that MR ligand binding domain (LBD) interacts strongly with only a few specific coactivator peptides in the presence of the agonist aldosterone and that these interactions are blocked by the antagonist eplerenone. We also discovered that cortisol, the preferred physiological ligand for the glucocorticoid receptor in humans, is a partial MR agonist/antagonist, providing a possible molecular explanation of the tissue-selective effects of glucocorticoids on MR. However, when we examined the coactivator and corepressor peptide interactions in the presence of cortisol, we found that MR bound with cortisol or aldosterone interacted with the same set of peptides. Thus, the partial agonism shown by cortisol is unlikely to be the result of differential interaction with known coactivators and corepressors. On the other hand, we have identified coactivator binding groove mutations that are critical for cortisol activation but not for aldosterone activation, suggesting that the two steroids induce different MR LBD conformations. In addition, we also show that cortisol becomes full agonist when S810L mutation is introduced in the LBD of MR. Interestingly, MR antagonists, such as eplerenone and progesterone, become partial agonist/antagonist of S810L but are still able to recruit LXXLL peptides to the mutant receptor. Together, these findings suggest a model to explain the MR activation by various ligands.


Asunto(s)
Regulación de la Expresión Génica , Receptores de Mineralocorticoides/metabolismo , Aldosterona/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Línea Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Eplerenona , Transferencia Resonante de Energía de Fluorescencia , Glucocorticoides/metabolismo , Humanos , Hidrocortisona/química , Hidrocortisona/metabolismo , Ligandos , Modelos Biológicos , Datos de Secuencia Molecular , Mutación , Péptidos/química , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Espironolactona/análogos & derivados , Espironolactona/química , Transcripción Genética , Transfección
14.
Biochim Biophys Acta ; 1598(1-2): 10-23, 2002 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-12147339

RESUMEN

MMP-2 is a member of the matrix metalloproteinase family that has been implicated in tumor cell metastasis and angiogenesis. Here, we describe the solution structure of a catalytic domain of MMP-2 complexed with a hydroxamic acid inhibitor (SC-74020), determined by three-dimensional heteronuclear NMR spectroscopy. The catalytic domain, designated MMP-2C, has a short peptide linker replacing the internal fibronectin-domain insertion and is enzymatically active. Distance geometry-simulated annealing calculations yielded 14 converged structures with atomic root-mean-square deviations (r.m.s.d.) of 1.02 and 1.62 A from the mean coordinate positions for the backbone and for all heavy atoms, respectively, when 11 residues at the N-terminus are excluded. The structure has the same global fold as observed for other MMP catalytic domains and is similar to previously solved crystal structures of MMP-2. Differences observed between the solution and the crystal structures, near the bottom of the S1' specificity loop, appear to be induced by the large inhibitor present in the solution structure. The MMP-2C solution structure is compared with MMP-8 crystal structure bound to the same inhibitor to highlight the differences especially in the S1' specificity loop. The finding provides a structural explanation for the selectivity between MMP-2 and MMP-8 that is achieved by large inhibitors.


Asunto(s)
Ácidos Hidroxámicos/química , Ácidos Hidroxámicos/farmacología , Metaloproteinasa 2 de la Matriz/metabolismo , Inhibidores de Proteasas/metabolismo , Sulfonamidas/química , Sulfonamidas/farmacología , Secuencia de Aminoácidos , Sitios de Unión , Dominio Catalítico , Humanos , Ácidos Hidroxámicos/síntesis química , Espectroscopía de Resonancia Magnética , Metaloproteinasa 2 de la Matriz/química , Modelos Moleculares , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/química , Conformación Proteica , Sulfonamidas/síntesis química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...