Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Endocrinol (Lausanne) ; 14: 1285269, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37941906

RESUMEN

Introduction: Polycystic ovary syndrome (PCOS) seems to be associated with increased ovarian sympathetic nerve activity and in rodent models of PCOS reducing the sympathetic drive to the ovary, through denervation or neuromodulation, improves ovulation rate. We hypothesised that sympathetic nerves work with gonadotropins to promote development and survival of small antral follicles to develop a polycystic ovary phenotype. Methods: Using a clinically realistic ovine model we showed a rich sympathetic innervation to the normal ovary and reinnervation after ovarian transplantation. Using needlepoint diathermy to the nerve plexus in the ovarian vascular pedicle we were able to denervate the ovary resulting in reduced intraovarian noradrenaline and tyrosine hydroxylase immunostained sympathetic nerves. We developed an acute polycystic ovary (PCO) model using gonadotrophin releasing hormone (GnRH) agonist followed infusion of follicle stimulating hormone (FSH) with increased pulsatile luteinising hormone (LH). This resulted in increased numbers of smaller antral follicles in the ovary when compared to FSH infusion suggesting a polycystic ovary. Results: Denervation had no effect of the survival or numbers of follicles in the acute PCO model and did not impact on ovulation, follicular and luteal hormone profiles in a normal cycle. Discussion: Although the ovary is richly inervated we did not find evidence for a role of sympathetic nerves in ovarian function or small follicle growth and survival.


Asunto(s)
Síndrome del Ovario Poliquístico , Femenino , Humanos , Ovinos , Animales , Síndrome del Ovario Poliquístico/complicaciones , Hormona Folículo Estimulante , Gonadotropinas , Oveja Doméstica , Desnervación
2.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-33972441

RESUMEN

Neuromodulation of immune function by stimulating the autonomic connections to the spleen has been demonstrated in rodent models. Consequently, neuroimmune modulation has been proposed as a new therapeutic strategy for the treatment of inflammatory conditions. However, demonstration of the translation of these immunomodulatory mechanisms in anatomically and physiologically relevant models is still lacking. Additionally, translational models are required to identify stimulation parameters that can be transferred to clinical applications of bioelectronic medicines. Here, we performed neuroanatomical and functional comparison of the mouse, rat, pig, and human splenic nerve using in vivo and ex vivo preparations. The pig was identified as a more suitable model of the human splenic innervation. Using functional electrophysiology, we developed a clinically relevant marker of splenic nerve engagement through stimulation-dependent reversible reduction in local blood flow. Translation of immunomodulatory mechanisms were then assessed using pig splenocytes and two models of acute inflammation in anesthetized pigs. The pig splenic nerve was shown to locally release noradrenaline upon stimulation, which was able to modulate cytokine production by pig splenocytes. Splenic nerve stimulation was found to promote cardiovascular protection as well as cytokine modulation in a high- and a low-dose lipopolysaccharide model, respectively. Importantly, splenic nerve-induced cytokine modulation was reproduced by stimulating the efferent trunk of the cervical vagus nerve. This work demonstrates that immune responses can be modulated by stimulation of spleen-targeted autonomic nerves in translational species and identifies splenic nerve stimulation parameters and biomarkers that are directly applicable to humans due to anatomical and electrophysiological similarities.


Asunto(s)
Sistema Inmunológico/inervación , Inmunomodulación/efectos de los fármacos , Bazo/inmunología , Sistema Nervioso Simpático/inmunología , Nervio Vago/inmunología , Animales , Femenino , Expresión Génica , Humanos , Sistema Inmunológico/efectos de los fármacos , Inflamación , Interleucina-6/genética , Interleucina-6/inmunología , Lipopolisacáridos/farmacología , Ratones , Microcirculación/efectos de los fármacos , Microcirculación/genética , Microcirculación/inmunología , Norepinefrina/farmacología , Ratas , Especificidad de la Especie , Bazo/efectos de los fármacos , Bazo/inervación , Bazo/patología , Porcinos , Sistema Nervioso Simpático/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Nervio Vago/efectos de los fármacos , Estimulación del Nervio Vago/métodos
3.
Commun Biol ; 3(1): 577, 2020 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-33067560

RESUMEN

Neuromodulation is a new therapeutic pathway to treat inflammatory conditions by modulating the electrical signalling pattern of the autonomic connections to the spleen. However, targeting this sub-division of the nervous system presents specific challenges in translating nerve stimulation parameters. Firstly, autonomic nerves are typically embedded non-uniformly among visceral and connective tissues with complex interfacing requirements. Secondly, these nerves contain axons with populations of varying phenotypes leading to complexities for axon engagement and activation. Thirdly, clinical translational of methodologies attained using preclinical animal models are limited due to heterogeneity of the intra- and inter-species comparative anatomy and physiology. Here we demonstrate how this can be accomplished by the use of in silico modelling of target anatomy, and validation of these estimations through ex vivo human tissue electrophysiology studies. Neuroelectrical models are developed to address the challenges in translation of parameters, which provides strong input criteria for device design and dose selection prior to a first-in-human trial.


Asunto(s)
Estimulación Eléctrica , Bazo/inervación , Animales , Estimulación Eléctrica/métodos , Terapia por Estimulación Eléctrica/métodos , Fenómenos Electrofisiológicos , Humanos , Bazo/anatomía & histología , Bazo/irrigación sanguínea , Bazo/citología , Porcinos
4.
Front Physiol ; 10: 349, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31024331

RESUMEN

Sympathetic innervation of the ovary in rodents occurs via two routes: the superior ovarian nerve (SON), which runs along the ovarian ligament, and the plexus nerve (PN), which is mainly associated with the vasculature. SON and ovarian norepinephrine (NE) levels play a major role in regulating ovarian cystic health. Although it was previously described that the polycystic ovarian phenotype (PCO) is causally related to hyperstimulation of the sympathetic nerves of the ovary, much less is known, however, regarding the role of PN in ovarian physiology. We studied the role of SON and PN in relation to the maintenance of the PCO phenotype induced in the rat by exposure to estradiol valerate (EV). EV exposure at 24 days old (juvenile exposure) increases NE in the ovary for up to 90 days after EV injection. SON or PN denervation (SONX and PNX) decreased NE. SONXreversed the acyclic condition from 30 days after surgery (p < 0.05), but PNXdid not. SONX was more effective than PNX to downregulate the increased number of cysts induced by EV, with the presence of the corpora lutea (CL, signifying ovulation) in the SONX group. Seventy percent of SONX rats presented with pregnancy at 60 days post-EV (6 of the 7 sperm-positive rats were pregnant); however, SONX rats had a reduced number (half) of pups compared with vehicle-treated rats and 60% more pups than EV rats. These data suggest that the SON plays a predominant role in follicular development, ovulation and pregnancy during ovarian diseases.

5.
Antiviral Res ; 154: 158-165, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29653131

RESUMEN

The DNA papillomaviruses infect squamous epithelium and can cause persistent, benign and sometimes malignant hyperproliferative lesions. Effective antiviral drugs to treat human papillomavirus (HPV) infection are lacking and here we investigate the anti-papillomavirus activity of novel epigenetic targeting drugs, BET bromodomain inhibitors. Bromodomain and Extra-Terminal domain (BET) proteins are host proteins which regulate gene transcription, they bind acetylated lysine residues in histones and non-histone proteins via bromodomains, functioning as scaffold proteins in the formation of transcriptional complexes at gene regulatory regions. The BET protein BRD4 has been shown to be involved in the papillomavirus life cycle, as a co-factor for viral E2 and also mediating viral partitioning in some virus types. We set out to study the activity of small molecule BET bromodomain inhibitors in models of papillomavirus infection. Several BET inhibitors reduced HPV11 E1ˆE4 mRNA expression in vitro and topical therapeutic administration of an exemplar compound I-BET762, abrogated CRPV cutaneous wart growth in rabbits, demonstrating translation of anti-viral effects to efficacy in vivo. Additionally I-BET762 markedly reduced viability of HPV16 infected W12 cells compared to non-infected C33A cells. The molecular mechanism for the cytotoxicity to W12 cells is unknown but may be through blocking viral-dependent cell-survival factors. We conclude that these effects, across multiple papillomavirus types and in vivo, highlight the potential to target BET bromodomains to treat HPV infection.


Asunto(s)
Benzodiazepinas/uso terapéutico , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Proteínas Nucleares/antagonistas & inhibidores , Papillomaviridae/efectos de los fármacos , Factores de Transcripción/antagonistas & inhibidores , Verrugas/tratamiento farmacológico , Acetilación , Animales , Línea Celular Tumoral , Supervivencia Celular , Epigénesis Genética , Lisina , Masculino , Papillomaviridae/genética , Dominios Proteicos , Conejos , Verrugas/virología
6.
J Med Chem ; 59(3): 1003-20, 2016 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-26751273

RESUMEN

FTY720 is the first oral small molecule approved for the treatment of people suffering from relapsing-remitting multiple sclerosis. It is a potent agonist of the S1P1 receptor, but its lack of selectivity against the S1P3 receptor has been linked to most of the cardiovascular side effects observed in the clinic. These findings have triggered intensive efforts toward the identification of a second generation of S1P3-sparing S1P1 agonists. We have recently disclosed a series of orally active tetrahydroisoquinoline (THIQ) compounds matching these criteria. In this paper we describe how we defined and implemented a strategy aiming at the discovery of selective structurally distinct follow-up agonists. This effort culminated with the identification of a series of orally active tetrahydropyrazolopyridines.


Asunto(s)
Descubrimiento de Drogas , Pirazoles/administración & dosificación , Pirazoles/farmacología , Piridinas/administración & dosificación , Piridinas/farmacología , Receptores de Lisoesfingolípidos/agonistas , Administración Oral , Animales , Línea Celular , Perros , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Ratones , Ratones Endogámicos , Estructura Molecular , Pirazoles/síntesis química , Pirazoles/química , Piridinas/síntesis química , Piridinas/química , Ratas , Ratas Endogámicas Lew , Ratas Sprague-Dawley , Receptores de Esfingosina-1-Fosfato , Relación Estructura-Actividad
7.
J Med Chem ; 58(20): 8236-56, 2015 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-26393276

RESUMEN

This article describes the finding of substantial upregulation of mRNA and enzymes of the cytochrome P450 1A family during a lead optimization campaign for small molecule S1P1 agonists. Fold changes in mRNA up to 10,000-fold for CYP1A1 in vivo in rat and cynomolgus monkey and up to 45-fold for CYP1A1 and CYP1A2 in vitro in rat and human hepatocytes were observed. Challenges observed with correlating induction in vitro and induction in vivo resulted in the implementation of a short, 4 day in vivo screening study in the rat which successfully identified noninducers. Subtle structure-activity relationships in this series of S1P1 agonists are described extending beyond planarity and lipophilicity, and the impact and considerations of AhR and CYP1A induction in the context of drug development are discussed.


Asunto(s)
Citocromo P-450 CYP1A1/biosíntesis , Citocromo P-450 CYP1A2/biosíntesis , Inducción Enzimática/efectos de los fármacos , Receptores de Hidrocarburo de Aril/agonistas , Receptores de Lisoesfingolípidos/agonistas , Animales , Perros , Diseño de Fármacos , Descubrimiento de Drogas , Hepatocitos/efectos de los fármacos , Humanos , Lípidos/química , Macaca fascicularis , Ratones , Modelos Moleculares , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Regulación hacia Arriba/efectos de los fármacos
8.
Nat Commun ; 5: 5418, 2014 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-25391636

RESUMEN

Emerging evidence suggests that RANKL-induced changes in chromatin state are important for osteoclastogenesis, but these epigenetic mechanisms are not well understood and have not been therapeutically targeted. In this study, we find that the small molecule I-BET151 that targets bromo and extra-terminal (BET) proteins that 'read' chromatin states by binding to acetylated histones strongly suppresses osteoclastogenesis. I-BET151 suppresses pathologic bone loss in TNF-induced inflammatory osteolysis, inflammatory arthritis and post-ovariectomy models. Transcriptome analysis identifies a MYC-NFAT axis important for osteoclastogenesis. Mechanistically, I-BET151 inhibits expression of the master osteoclast regulator NFATC1 by suppressing expression and recruitment of its newly identified upstream regulator MYC. MYC is elevated in rheumatoid arthritis macrophages and its induction by RANKL is important for osteoclastogenesis and TNF-induced bone resorption. These findings highlight the importance of an I-BET151-inhibited MYC-NFAT axis in osteoclastogenesis, and suggest targeting epigenetic chromatin regulators holds promise for treatment of inflammatory and oestrogen deficiency-mediated pathologic bone resorption.


Asunto(s)
Resorción Ósea/fisiopatología , Epigénesis Genética/fisiología , Inflamación/fisiopatología , Osteoclastos/fisiología , Osteogénesis/fisiología , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Femenino , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/antagonistas & inhibidores , Factores de Transcripción NFATC/fisiología , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteoporosis/fisiopatología , Ovariectomía , Ligando RANK/fisiología
9.
J Med Chem ; 57(24): 10424-42, 2014 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-25431977

RESUMEN

The efficacy of the recently approved drug fingolimod (FTY720) in multiple sclerosis patients results from the action of its phosphate metabolite on sphingosine-1-phosphate S1P1 receptors, while a variety of side effects have been ascribed to its S1P3 receptor activity. Although S1P and phospho-fingolimod share the same structural elements of a zwitterionic headgroup and lipophilic tail, a variety of chemotypes have been found to show S1P1 receptor agonism. Here we describe a study of the tolerance of the S1P1 and S1P3 receptors toward bicyclic heterocycles of systematically varied shape and connectivity incorporating acidic, basic, or zwitterionic headgroups. We compare their physicochemical properties, their performance in in vitro and in vivo pharmacokinetic models, and their efficacy in peripheral lymphocyte lowering. The campaign resulted in the identification of several potent S1P1 receptor agonists with good selectivity vs S1P3 receptors, efficacy at <1 mg/kg oral doses, and developability properties suitable for progression into preclinical development.


Asunto(s)
Ácidos Carboxílicos/química , Compuestos Heterocíclicos/química , Inmunosupresores/farmacología , Inmunosupresores/farmacocinética , Linfocitos/efectos de los fármacos , Receptores de Lisoesfingolípidos/agonistas , Animales , Células Cultivadas , Inmunosupresores/síntesis química , Linfocitos/citología , Masculino , Modelos Moleculares , Estructura Molecular , Ratas Endogámicas Lew , Relación Estructura-Actividad
10.
ChemMedChem ; 9(3): 580-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24000170

RESUMEN

Bromodomains (BRDs) are small protein domains found in a variety of proteins that recognize and bind to acetylated histone tails. This binding affects chromatin structure and facilitates the localisation of transcriptional complexes to specific genes, thereby regulating epigenetically controlled processes including gene transcription and mRNA elongation. Inhibitors of the bromodomain and extra-terminal (BET) proteins BRD2-4 and T, which prevent bromodomain binding to acetyl-modified histone tails, have shown therapeutic promise in several diseases. We report here the discovery of 1,5-naphthyridine derivatives as potent inhibitors of the BET bromodomain family with good cell activity and oral pharmacokinetic parameters. X-ray crystal structures of naphthyridine isomers have been solved and quantum mechanical calculations have been used to explain the higher affinity of the 1,5-isomer over the others. The best compounds were progressed in a mouse model of inflammation and exhibited dose-dependent anti-inflammatory pharmacology.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Naftiridinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Animales , Antiinflamatorios no Esteroideos/química , Proteínas Cromosómicas no Histona , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Histonas/química , Histonas/metabolismo , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Estructura Molecular , Naftiridinas/química , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína/efectos de los fármacos , Relación Estructura-Actividad , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo
11.
Blood ; 123(5): 697-705, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24335499

RESUMEN

The bromodomain and extraterminal (BET) protein BRD2-4 inhibitors hold therapeutic promise in preclinical models of hematologic malignancies. However, translation of these data to molecules suitable for clinical development has yet to be accomplished. Herein we expand the mechanistic understanding of BET inhibitors in multiple myeloma by using the chemical probe molecule I-BET151. I-BET151 induces apoptosis and exerts strong antiproliferative effect in vitro and in vivo. This is associated with contrasting effects on oncogenic MYC and HEXIM1, an inhibitor of the transcriptional activator P-TEFb. I-BET151 causes transcriptional repression of MYC and MYC-dependent programs by abrogating recruitment to the chromatin of the P-TEFb component CDK9 in a BRD2-4-dependent manner. In contrast, transcriptional upregulation of HEXIM1 is BRD2-4 independent. Finally, preclinical studies show that I-BET762 has a favorable pharmacologic profile as an oral agent and that it inhibits myeloma cell proliferation, resulting in survival advantage in a systemic myeloma xenograft model. These data provide a strong rationale for extending the clinical testing of the novel antimyeloma agent I-BET762 and reveal insights into biologic pathways required for myeloma cell proliferation.


Asunto(s)
Antineoplásicos/uso terapéutico , Benzodiazepinas/uso terapéutico , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Benzodiazepinas/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Ratones , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas de Unión al ARN/genética , Factores de Transcripción , Activación Transcripcional/efectos de los fármacos , Células Tumorales Cultivadas
12.
Bioorg Med Chem Lett ; 23(24): 6890-6, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24269482

RESUMEN

This Letter describes the discovery of GSK189254 and GSK239512 that were progressed as clinical candidates to explore the potential of H3 receptor antagonists as novel therapies for the treatment of Alzheimer's disease and other dementias. By carefully controlling the physicochemical properties of the benzazepine series and through the implementation of an aggressive and innovative screening strategy that employed high throughput in vivo assays to efficiently triage compounds, the medicinal chemistry effort was able to rapidly progress the benzazepine class of H3 antagonists through to the identification of clinical candidates with robust in vivo efficacy and excellent developability properties.


Asunto(s)
Benzazepinas/química , Antagonistas de los Receptores Histamínicos H3/química , Receptores Histamínicos H3/química , Animales , Benzazepinas/farmacocinética , Perros , Semivida , Haplorrinos , Antagonistas de los Receptores Histamínicos H3/síntesis química , Antagonistas de los Receptores Histamínicos H3/farmacocinética , Humanos , Masculino , Microsomas Hepáticos/metabolismo , Niacinamida/análogos & derivados , Niacinamida/química , Niacinamida/farmacocinética , Unión Proteica , Ratas , Ratas Sprague-Dawley , Receptores Histamínicos H3/metabolismo , Relación Estructura-Actividad
13.
Bioorg Med Chem Lett ; 23(24): 6897-901, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24161834

RESUMEN

This Letter describes the discovery of a novel series of H3 receptor antagonists. The initial medicinal chemistry strategy focused on deconstructing and simplifying an early screening hit which rapidly led to the discovery of a novel series of H3 receptor antagonists based on the benzazepine core. Employing an H3 driven pharmacodynamic model, the series was then further optimised through to a lead compound that showed robust in vivo functional activity and possessed overall excellent developability properties.


Asunto(s)
Benzazepinas/química , Antagonistas de los Receptores Histamínicos H3/química , Receptores Histamínicos H3/química , Animales , Benzazepinas/síntesis química , Benzazepinas/farmacocinética , Citocromo P-450 CYP2D6/química , Citocromo P-450 CYP2D6/metabolismo , Evaluación Preclínica de Medicamentos , Semivida , Antagonistas de los Receptores Histamínicos H3/síntesis química , Antagonistas de los Receptores Histamínicos H3/farmacocinética , Humanos , Microsomas Hepáticos/metabolismo , Unión Proteica , Ratas , Receptores Histamínicos H3/genética , Receptores Histamínicos H3/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Relación Estructura-Actividad
14.
J Med Chem ; 56(19): 7501-15, 2013 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-24015967

RESUMEN

The bromo and extra C-terminal domain (BET) family of bromodomains are involved in binding epigenetic marks on histone proteins, more specifically acetylated lysine residues. This paper describes the discovery and structure-activity relationships (SAR) of potent benzodiazepine inhibitors that disrupt the function of the BET family of bromodomains (BRD2, BRD3, and BRD4). This work has yielded a potent, selective compound I-BET762 that is now under evaluation in a phase I/II clinical trial for nuclear protein in testis (NUT) midline carcinoma and other cancers.


Asunto(s)
Antineoplásicos/farmacología , Benzodiazepinas/farmacología , Proteínas Nucleares/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Animales , Antiinflamatorios/síntesis química , Antiinflamatorios/farmacocinética , Antiinflamatorios/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/farmacocinética , Apolipoproteína A-I/biosíntesis , Benzodiazepinas/síntesis química , Benzodiazepinas/farmacocinética , Proteínas de Ciclo Celular , Perros , Epigénesis Genética , Humanos , Macaca fascicularis , Ratones , Modelos Moleculares , Permeabilidad , Estructura Terciaria de Proteína , Ratas , Estereoisomerismo , Relación Estructura-Actividad
15.
Proc Natl Acad Sci U S A ; 109(36): 14532-7, 2012 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-22912406

RESUMEN

Bromodomain-containing proteins bind acetylated lysine residues on histone tails and are involved in the recruitment of additional factors that mediate histone modifications and enable transcription. A compound, I-BET-762, that inhibits binding of an acetylated histone peptide to proteins of the bromodomain and extra-terminal domain (BET) family, was previously shown to suppress the production of proinflammatory proteins by macrophages and block acute inflammation in mice. Here, we investigated the effect of short-term treatment with I-BET-762 on T-cell function. Treatment of naïve CD4(+) T cells with I-BET-762 during the first 2 d of differentiation had long-lasting effects on subsequent gene expression and cytokine production. Gene expression analysis revealed up-regulated expression of several antiinflammatory gene products, including IL-10, Lag3, and Egr2, and down-regulated expression of several proinflammatory cytokines including GM-CSF and IL-17. The short 2-d treatment with I-BET-762 inhibited the ability of antigen-specific T cells, differentiated under Th1 but not Th17 conditions in vitro, to induce pathogenesis in an adoptive transfer model of experimental autoimmune encephalomyelitis. The suppressive effects of I-BET-762 on T-cell mediated inflammation in vivo were accompanied by decreased recruitment of macrophages, consistent with decreased GM-CSF production by CNS-infiltrating T cells. These effects were mimicked by an inhibitor of c-myc function, implicating reduced expression of c-myc and GM-CSF as one avenue by which I-BET-762 suppresses the inflammatory functions of T cells. Our study demonstrates that inhibiting the functions of BET-family proteins during early T-cell differentiation causes long-lasting suppression of the proinflammatory functions of Th1 cells.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Regulación de la Expresión Génica/inmunología , Proteínas Nucleares/inmunología , alfa-Amilasas Salivales/antagonistas & inhibidores , Factores de Transcripción/inmunología , Transcripción Genética/inmunología , Traslado Adoptivo , Animales , Benzodiazepinas/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Histonas/metabolismo , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices , Proteínas Nucleares/metabolismo , Fosforilación , Factor B de Elongación Transcripcional Positiva/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tiazoles/farmacología , Factores de Transcripción/metabolismo
16.
Bioorg Med Chem Lett ; 22(8): 2968-72, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22437115

RESUMEN

A novel series of quinoline isoxazole BET family bromodomain inhibitors are discussed. Crystallography is used to illustrate binding modes and rationalize their SAR. One member, I-BET151 (GSK1210151A), shows good oral bioavailability in both the rat and minipig as well as demonstrating efficient suppression of bacterial induced inflammation and sepsis in a murine in vivo endotoxaemia model.


Asunto(s)
Compuestos Heterocíclicos de 4 o más Anillos/química , Isoxazoles/síntesis química , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Quinolinas/síntesis química , Animales , Sitios de Unión , Cristalografía por Rayos X , Cobayas , Compuestos Heterocíclicos de 4 o más Anillos/metabolismo , Inflamación/tratamiento farmacológico , Isoxazoles/química , Isoxazoles/farmacología , Ratones , Modelos Moleculares , Unión Proteica/efectos de los fármacos , Quinolinas/química , Quinolinas/farmacología , Ratas
17.
Xenobiotica ; 42(7): 671-86, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22225501

RESUMEN

Sphingosine-1-phosphate (S1P(1)) receptor agonists such as Fingolimod (FTY-720) are a novel class of immunomodulators that have clinical utility in the treatment of remitting relapsing multiples sclerosis. This class of compound act by inducing peripheral lymphopenia. Using an integrated pharmacokinetic/pharmacodynamic (PK-PD) approach based on an in vivo rat model, novel S1P(1) agonists were identified with a predicted more rapid rate of reversibility of lymphocyte reduction in human compared to Fingolimod. The in vivo potency of 15 compounds based on PK-PD modelling of the rat lymphocyte reduction model was correlated with in vitro measures of potency at the S1P(1) receptor using ß arrestin recruitment and G-protein signalling. A structurally novel S1P(1) agonist was identified and predictions of human pharmacokinetics and clinical dose are presented.


Asunto(s)
Lisofosfolípidos/agonistas , Glicoles de Propileno/farmacocinética , Esfingosina/análogos & derivados , Animales , Arrestina/metabolismo , Clorhidrato de Fingolimod , Proteínas de Unión al GTP/metabolismo , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Lisofosfolípidos/metabolismo , Masculino , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Glicoles de Propileno/farmacología , Glicoles de Propileno/uso terapéutico , Ratas , Ratas Endogámicas , Transducción de Señal , Esfingosina/agonistas , Esfingosina/metabolismo , Esfingosina/farmacocinética , Esfingosina/farmacología , Esfingosina/uso terapéutico
18.
J Biomol Screen ; 16(10): 1170-85, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21956175

RESUMEN

Bromodomains are structurally conserved protein modules present in a large number of chromatin-associated proteins and in many nuclear histone acetyltransferases. The bromodomain functions as an acetyl-lysine binding domain and has been shown to be pivotal in regulating protein-protein interactions in chromatin-mediated cellular gene transcription, cell proliferation, and viral transcriptional activation. Structural analyses of these modules in complex with acetyl-lysine peptide ligands provide insights into the molecular basis for recognition and ligand selectivity within this epigenetic reader family. However, there are significant challenges in configuring assays to identify inhibitors of these proteins. This review focuses on the progress made in developing methods to identify peptidic and small-molecule ligands using biophysical label-free and biochemical approaches. The advantage of each technique and the results reported are summarized, highlighting the potential applicably to other reader domains and the caveats in translation from simple in vitro systems to a biological context.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Histona Acetiltransferasas/metabolismo , Histonas/metabolismo , Animales , Descubrimiento de Drogas/métodos , Histona Acetiltransferasas/química , Humanos , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína
19.
J Med Chem ; 54(19): 6724-33, 2011 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-21838322

RESUMEN

2-Amino-2-(4-octylphenethyl)propane-1,3-diol 1 (fingolimod, FTY720) has been recently marketed in the United States for the treatment of patients with remitting relapsing multiple sclerosis (RRMS). Its efficacy has been primarily linked to the agonism on T cells of S1P(1), one of the five sphingosine 1-phosphate (S1P) G-protein-coupled receptors, while its cardiovascular side effects have been associated with activity at S1P(3). Emerging data suggest that the ability of this molecule to cross the blood-brain barrier and to interact with both S1P(1) and S1P(5) in the central nervous system (CNS) may contribute to its efficacy in treating patients with RRMS. We have recently disclosed the structure of an advanced, first generation S1P(3)-sparing S1P(1) agonist, a zwitterion with limited CNS exposure. In this Article, we highlight our strategy toward the identification of CNS-penetrant S1P(3)-sparing S1P(1) and S1P(5) agonists resulting in the discovery of 5-(3-{2-[2-hydroxy-1-(hydroxymethyl)ethyl]-5-methyl-1,2,3,4-tetrahydro-6-isoquinolinyl}-1,2,4-oxadiazol-5-yl)-2-[(1-methylethyl)oxy]benzonitrile 15. Its exceptional in vivo potency and good pharmacokinetic properties translate into a very low predicted therapeutic dose in human (<1 mg p.o. once daily).


Asunto(s)
Azepinas/síntesis química , Encéfalo/metabolismo , Isoquinolinas/síntesis química , Oxadiazoles/síntesis química , Receptores de Lisoesfingolípidos/agonistas , Administración Oral , Animales , Azepinas/farmacocinética , Azepinas/farmacología , Disponibilidad Biológica , Barrera Hematoencefálica/metabolismo , Línea Celular , Permeabilidad de la Membrana Celular , Perros , Isoquinolinas/farmacocinética , Isoquinolinas/farmacología , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Oxadiazoles/farmacocinética , Oxadiazoles/farmacología , Ratas , Receptores de Lisoesfingolípidos/metabolismo , Solubilidad
20.
ACS Med Chem Lett ; 2(6): 444-9, 2011 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-24900328

RESUMEN

Gilenya (fingolimod, FTY720) was recently approved by the U.S. FDA for the treatment of patients with remitting relapsing multiple sclerosis (RRMS). It is a potent agonist of four of the five sphingosine 1-phosphate (S1P) G-protein-coupled receptors (S1P1 and S1P3-5). It has been postulated that fingolimod's efficacy is due to S1P1 agonism, while its cardiovascular side effects (transient bradycardia and hypertension) are due to S1P3 agonism. We have discovered a series of selective S1P1 agonists, which includes 3-[6-(5-{3-cyano-4-[(1-methylethyl)oxy]phenyl}-1,2,4-oxadiazol-3-yl)-5-methyl-3,4-dihydro-2(1H)-isoquinolinyl]propanoate, 20, a potent, S1P3-sparing, orally active S1P1 agonist. Compound 20 is as efficacious as fingolimod in a collagen-induced arthritis model and shows excellent pharmacokinetic properties preclinically. Importantly, the selectivity of 20 against S1P3 is responsible for an absence of cardiovascular signal in telemetered rats, even at high dose levels.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA