Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Pharmacol Toxicol ; 63: 295-320, 2023 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-36662583

RESUMEN

The actions of estrogens and related estrogenic molecules are complex and multifaceted in both sexes. A wide array of natural, synthetic, and therapeutic molecules target pathways that produce and respond to estrogens. Multiple receptors promulgate these responses, including the classical estrogen receptors of the nuclear hormone receptor family (estrogen receptors α and ß), which function largely as ligand-activated transcription factors, and the 7-transmembrane G protein-coupled estrogen receptor, GPER, which activates a diverse array of signaling pathways. The pharmacology and functional roles of GPER in physiology and disease reveal important roles in responses to both natural and synthetic estrogenic compounds in numerous physiological systems. These functions have implications in the treatment of myriad disease states, including cancer, cardiovascular diseases, and metabolic disorders. This review focuses on the complex pharmacology of GPER and summarizes major physiological functions of GPER and the therapeutic implications and ongoing applications of GPER-targeted compounds.


Asunto(s)
Estrógenos , Receptores de Estrógenos , Masculino , Femenino , Humanos , Receptores de Estrógenos/metabolismo , Estrógenos/farmacología , Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Proteínas de Unión al GTP/metabolismo
2.
Pharmacol Rev ; 67(3): 505-40, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26023144

RESUMEN

Estrogens are critical mediators of multiple and diverse physiologic effects throughout the body in both sexes, including the reproductive, cardiovascular, endocrine, nervous, and immune systems. As such, alterations in estrogen function play important roles in many diseases and pathophysiological conditions (including cancer), exemplified by the lower prevalence of many diseases in premenopausal women. Estrogens mediate their effects through multiple cellular receptors, including the nuclear receptor family (ERα and ERß) and the G protein-coupled receptor (GPCR) family (GPR30/G protein-coupled estrogen receptor [GPER]). Although both receptor families can initiate rapid cell signaling and transcriptional regulation, the nuclear receptors are traditionally associated with regulating gene expression, whereas GPCRs are recognized as mediating rapid cellular signaling. Estrogen-activated pathways are not only the target of multiple therapeutic agents (e.g., tamoxifen, fulvestrant, raloxifene, and aromatase inhibitors) but are also affected by a plethora of phyto- and xeno-estrogens (e.g., genistein, coumestrol, bisphenol A, dichlorodiphenyltrichloroethane). Because of the existence of multiple estrogen receptors with overlapping ligand specificities, expression patterns, and signaling pathways, the roles of the individual receptors with respect to the diverse array of endogenous and exogenous ligands have been challenging to ascertain. The identification of GPER-selective ligands however has led to a much greater understanding of the roles of this receptor in normal physiology and disease as well as its interactions with the classic estrogen receptors ERα and ERß and their signaling pathways. In this review, we describe the history and characterization of GPER over the past 15 years focusing on the pharmacology of steroidal and nonsteroidal compounds that have been employed to unravel the biology of this most recently recognized estrogen receptor.


Asunto(s)
Estrógenos/metabolismo , Receptores de Estrógenos/efectos de los fármacos , Receptores Acoplados a Proteínas G/efectos de los fármacos , Animales , Femenino , Humanos , Ligandos , Masculino , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Esteroides/farmacología
3.
J Physiol ; 591(17): 4223-35, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23878371

RESUMEN

In addition to acting on mineralocorticoid receptors, aldosterone has been recently shown to activate the G protein-coupled oestrogen receptor (GPER) in vascular cells. In light of the newly identified role for GPER in vagal cardiac control, we examined whether or not aldosterone activates GPER in rat nucleus ambiguus. Aldosterone produced a dose-dependent increase in cytosolic Ca(2+) concentration in retrogradely labelled cardiac vagal neurons of nucleus ambiguus; the response was abolished by pretreatment with the GPER antagonist G-36, but was not affected by the mineralocorticoid receptor antagonists, spironolactone and eplerenone. In Ca(2+)-free saline, the response to aldosterone was insensitive to blockade of the Ca(2+) release from lysosomes, while it was reduced by blocking the Ca(2+) release via ryanodine receptors and abolished by blocking the IP3 receptors. Aldosterone induced Ca(2+) influx via P/Q-type Ca(2+) channels, but not via L-type and N-type Ca(2+) channels. Aldosterone induced depolarization of cardiac vagal neurons of nucleus ambiguus that was sensitive to antagonism of GPER but not of mineralocorticoid receptor. in vivo studies, using telemetric measurement of heart rate, indicate that microinjection of aldosterone into the nucleus ambiguus produced a dose-dependent bradycardia in conscious, freely moving rats. Aldosterone-induced bradycardia was blocked by the GPER antagonist, but not by the mineralocorticoid receptor antagonists. In summary, we report for the first time that aldosterone decreases heart rate by activating GPER in cardiac vagal neurons of nucleus ambiguus.


Asunto(s)
Aldosterona/farmacología , Corazón/fisiología , Receptores de Estrógenos/metabolismo , Nervio Vago/fisiología , Potenciales de Acción , Animales , Calcio/metabolismo , Señalización del Calcio , Células Cultivadas , Corazón/efectos de los fármacos , Corazón/inervación , Frecuencia Cardíaca , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/fisiología , Ratas , Ratas Sprague-Dawley , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Mineralocorticoides/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Nervio Vago/efectos de los fármacos , Nervio Vago/metabolismo
4.
Exp Physiol ; 98(3): 679-91, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23104934

RESUMEN

The G protein-coupled estrogen receptor (GPER) has been identified in several brain regions, including cholinergic neurons of the nucleus ambiguus, which are critical for parasympathetic cardiac regulation. Using calcium imaging and electrophysiological techniques, microinjection into the nucleus ambiguus and blood pressure measurement, we examined the in vitro and in vivo effects of GPER activation in nucleus ambiguus neurons. A GPER selective agonist, G-1, produced a sustained increase in cytosolic Ca(2+) concentration in a concentration-dependent manner in retrogradely labelled cardiac vagal neurons of nucleus ambiguus. The increase in cytosolic Ca(2+) produced by G-1 was abolished by pretreatment with G36, a GPER antagonist. G-1 depolarized cultured cardiac vagal neurons of the nucleus ambiguus. The excitatory effect of G-1 was also identified by whole-cell visual patch-clamp recordings in nucleus ambiguus neurons, in medullary slices. To validate the physiological relevance of our in vitro studies, we carried out in vivo experiments. Microinjection of G-1 into the nucleus ambiguus elicited a decrease in heart rate; the effect was blocked by prior microinjection of G36. Systemic injection of G-1, in addition to a previously reported decrease in blood pressure, also reduced the heart rate. The G-1-induced bradycardia was prevented by systemic injection of atropine, a muscarinic antagonist, or by bilateral microinjection of G36 into the nucleus ambiguus. Our results indicate that GPER-mediated bradycardia occurs via activation of cardiac parasympathetic neurons of the nucleus ambiguus and support the involvement of the GPER in the modulation of cardiac vagal tone.


Asunto(s)
Bradicardia/inducido químicamente , Bulbo Raquídeo/fisiología , Receptores de Estrógenos/fisiología , Receptores Acoplados a Proteínas G/fisiología , Animales , Calcio/metabolismo , Ciclopentanos/farmacología , Corazón/efectos de los fármacos , Corazón/fisiología , Masculino , Bulbo Raquídeo/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Sistema Nervioso Parasimpático/efectos de los fármacos , Sistema Nervioso Parasimpático/fisiología , Quinolinas/farmacología , Ratas , Ratas Wistar , Receptores de Estrógenos/agonistas , Receptores de Estrógenos/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Nervio Vago/efectos de los fármacos , Nervio Vago/fisiología
5.
Am J Physiol Cell Physiol ; 301(5): C1262-9, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21865584

RESUMEN

G protein-coupled estrogen receptor 1 (GPER), also named GPR30, has been previously identified in the female reproductive system. In this study, GPER expression was found in the female rat myometrium by reverse transcriptase-polymerase chain reaction and immunocytochemistry. Using GPER-selective ligands, we assessed the effects of the GPER activation on resting membrane potential and cytosolic Ca(2+) concentration ([Ca(2+)](i)) in rat myometrial cells, as well as on contractility of rat uterine strips. G-1, a specific GPER agonist, induced a concentration-dependent depolarization and increase in [Ca(2+)](i) in myometrial cells. The depolarization was abolished in Na(+)-free saline. G-1-induced [Ca(2+)](i) increase was markedly decreased by nifedipine, a L-type Ca(2+) channel blocker, by Ca(2+)-free or Na(+)-free saline. Intracellular administration of G-1 produced a faster and transitory increase in [Ca(2+)](i), with a higher amplitude than that induced by extracellular application, supporting an intracellular localization of the functional GPER in myometrial cells. Depletion of internal Ca(2+) stores with thapsigargin produced a robust store-activated Ca(2+) entry; the Ca(2+) response to G-1 was similar to the constitutive Ca(2+) entry and did not seem to involve store-operated Ca(2+) entry. In rat uterine strips, administration of G-1 increased the frequency and amplitude of contractions and the area under the contractility curve. The effects of G-1 on membrane potential, [Ca(2+)](i), and uterine contractility were prevented by pretreatment with G-15, a GPER antagonist, further supporting the involvement of GPER in these responses. Taken together, our results indicate that GPER is expressed and functional in rat myometrium. GPER activation produces depolarization, elevates [Ca(2+)](i) and increases contractility in myometrial cells.


Asunto(s)
Miometrio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Benzodioxoles/farmacología , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Ciclopentanos/farmacología , Inhibidores Enzimáticos/farmacología , Femenino , Potenciales de la Membrana/efectos de los fármacos , Nifedipino/farmacología , Quinolinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Tapsigargina/farmacología , Contracción Uterina/efectos de los fármacos , Contracción Uterina/metabolismo
6.
J Am Chem Soc ; 133(17): 6780-90, 2011 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-21473622

RESUMEN

A new class of fluorescent triazaborolopyridinium compounds was synthesized from hydrazones of 2-hydrazinylpyridine (HPY) and evaluated as potential dyes for live-cell imaging applications. The HPY dyes are small, their absorption/emission properties are tunable through variation of pyridyl or hydrazone substituents, and they offer favorable photophysical characteristics featuring large Stokes shifts and general insensitivity to solvent or pH. The stability, neutral charge, cell membrane permeability, and favorable relative influences on the water solubility of HPY conjugates are complementary to existing fluorescent dyes and offer advantages for the development of receptor-targeted small-molecule probes. This potential was assessed through the development of a new class of cysteine-derived HPY-conjugate imaging agents for the kinesin spindle protein (KSP) that is expressed in the cytoplasm during mitosis and is a promising chemotherapeutic target. Conjugates possessing the neutral HPY or charged Alexa Fluor dyes that function as potent, selective allosteric inhibitors of the KSP motor were compared using biochemical and cell-based phenotypic assays and live-cell imaging. These results demonstrate the effectiveness of the HPY dye moiety as a component of probes for an intracellular protein target and highlight the importance of dye structure in determining the pathway of cell entry and the overall performance of small-molecule conjugates as imaging agents.


Asunto(s)
Membrana Celular/metabolismo , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Compuestos de Piridinio/química , Compuestos de Piridinio/metabolismo , Permeabilidad de la Membrana Celular , Colorantes Fluorescentes/síntesis química , Células HeLa , Humanos , Compuestos de Piridinio/síntesis química , Piridonas/síntesis química , Piridonas/química
7.
Nat Chem Biol ; 5(6): 421-7, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19430488

RESUMEN

Estrogen is central to many physiological processes throughout the human body. We have previously shown that the G protein-coupled receptor GPR30 (also known as GPER), in addition to classical nuclear estrogen receptors (ER and ER), activates cellular signaling pathways in response to estrogen. In order to distinguish between the actions of classical estrogen receptors and GPR30, we have previously characterized G-1 (1), a selective agonist of GPR30. To complement the pharmacological properties of G-1, we sought to identify an antagonist of GPR30 that displays similar selectivity against the classical estrogen receptors. Here we describe the identification and characterization of G15 (2), a G-1 analog that binds to GPR30 with high affinity and acts as an antagonist of estrogen signaling through GPR30. In vivo administration of G15 revealed that GPR30 contributes to both uterine and neurological responses initiated by estrogen. The identification of this antagonist will accelerate the evaluation of the roles of GPR30 in human physiology.


Asunto(s)
Receptores de Estrógenos/fisiología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Animales , Células COS , Chlorocebus aethiops , Estrógenos/metabolismo , Femenino , Humanos , Ligandos , Masculino , Ratones , Ratones Endogámicos ICR , Resonancia Magnética Nuclear Biomolecular , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal
8.
Bioorg Med Chem ; 19(18): 5446-53, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21855351

RESUMEN

Assembly of a bipolar mitotic spindle requires the action of class 5 kinesins, and inhibition or depletion of this motor results in mitotic arrest and apoptosis. S-Trityl-l-cysteine is an allosteric inhibitor of vertebrate Kinesin Spindle Protein (KSP) that has generated considerable interest due to its anti-cancer properties, however, poor pharmacological properties have limited the use of this compound. We have modified the triphenylmethyl and cysteine groups, guided by biochemical and cell-based assays, to yield new cysteinol and cysteamine derivatives with increased inhibitory activity, greater efficacy in model systems, and significantly enhanced potency against the NCI60 tumor panel. These results reveal a promising new class of conformationally-flexible small molecules as allosteric KSP inhibitors for use as research tools, with activities that provide impetus for further development as anti-tumor agents.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Cisteamina/análogos & derivados , Cinesinas/antagonistas & inhibidores , Compuestos de Tritilo/farmacología , Animales , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Cisteamina/síntesis química , Cisteamina/química , Cisteamina/farmacología , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Embrión no Mamífero/efectos de los fármacos , Células HeLa , Humanos , Modelos Moleculares , Estructura Molecular , Erizos de Mar/efectos de los fármacos , Erizos de Mar/embriología , Estereoisomerismo , Relación Estructura-Actividad , Compuestos de Tritilo/síntesis química , Compuestos de Tritilo/química
9.
Virology ; 552: 94-106, 2021 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-33120225

RESUMEN

Drugs against flaviviruses such as dengue (DENV) and Zika (ZIKV) virus are urgently needed. We previously demonstrated that three fluoroquinolones, ciprofloxacin, enoxacin, and difloxacin, suppress replication of six flaviviruses. To investigate the barrier to resistance and mechanism(s) of action of these drugs, DENV-4 was passaged in triplicate in HEK-293 cells in the presence or absence of each drug. Resistance to ciprofloxacin was detected by the seventh passage and to difloxacin by the tenth, whereas resistance to enoxacin did not occur within ten passages. Two putative resistance-conferring mutations were detected in the envelope gene of ciprofloxacin and difloxacin-resistant DENV-4. In the absence of ciprofloxacin, ciprofloxacin-resistant viruses sustained a significantly higher viral titer than control viruses in HEK-293 and HuH-7 cells and resistant viruses were more stable than control viruses at 37 °C. These results suggest that the mechanism of action of ciprofloxacin and difloxacin involves interference with virus binding or entry.


Asunto(s)
Evolución Biológica , Virus del Dengue/efectos de los fármacos , Virus del Dengue/fisiología , Dengue/virología , Fluoroquinolonas/farmacología , Aptitud Genética/efectos de los fármacos , Fenómenos Fisiológicos de los Virus/efectos de los fármacos , Adaptación Biológica , Animales , Antivirales/farmacología , Línea Celular , Chlorocebus aethiops , Ciprofloxacina/análogos & derivados , Ciprofloxacina/farmacología , Farmacorresistencia Viral , Enoxacino/farmacología , Células HEK293 , Interacciones Microbiota-Huesped , Humanos , Mutación , Células Vero , Envoltura Viral/fisiología
10.
Org Biomol Chem ; 8(9): 2252-9, 2010 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-20401403

RESUMEN

The GPR30 agonist probe G-1 and structural analogs were efficiently synthesized using multicomponent or stepwise Sc(III)-catalyzed aza-Diels-Alder cyclization. Optimization of solvent and reaction temperature provided enhanced endo-diastereoselectivity.


Asunto(s)
Ciclopentanos/síntesis química , Ciclopentanos/farmacología , Quinolinas/síntesis química , Quinolinas/farmacología , Receptores Acoplados a Proteínas G/agonistas , Ciclopentanos/química , Estructura Molecular , Quinolinas/química , Estereoisomerismo , Relación Estructura-Actividad
11.
Sci Transl Med ; 12(528)2020 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-31996464

RESUMEN

Human obesity has become a global health epidemic, with few safe and effective pharmacological therapies currently available. The systemic loss of ovarian estradiol (E2) in women after menopause greatly increases the risk of obesity and metabolic dysfunction, revealing the critical role of E2 in this setting. The salutary effects of E2 are traditionally attributed to the classical estrogen receptors ERα and ERß, with the contribution of the G protein-coupled estrogen receptor (GPER) still largely unknown. Here, we used ovariectomy- and diet-induced obesity (DIO) mouse models to evaluate the preclinical activity of GPER-selective small-molecule agonist G-1 (also called Tespria) against obesity and metabolic dysfunction. G-1 treatment of ovariectomized female mice (a model of postmenopausal obesity) reduced body weight and improved glucose homeostasis without changes in food intake, fuel source usage, or locomotor activity. G-1-treated female mice also exhibited increased energy expenditure, lower body fat content, and reduced fasting cholesterol, glucose, insulin, and inflammatory markers but did not display feminizing effects on the uterus (imbibition) or beneficial effects on bone health. G-1 treatment of DIO male mice did not elicit weight loss but prevented further weight gain and improved glucose tolerance, indicating that G-1 improved glucose homeostasis independently of its antiobesity effects. However, in ovariectomized DIO female mice, G-1 continued to elicit weight loss, reflecting possible sex differences in the mechanisms of G-1 action. In conclusion, this work demonstrates that GPER-selective agonism is a viable therapeutic approach against obesity, diabetes, and associated metabolic abnormalities in multiple preclinical male and female models.


Asunto(s)
Diabetes Mellitus/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Receptores Acoplados a Proteínas G/agonistas , Tejido Adiposo/patología , Adiposidad/efectos de los fármacos , Animales , Respiración de la Célula , Modelos Animales de Enfermedad , Metabolismo Energético , Estrógenos/deficiencia , Femenino , Genes Mitocondriales , Glucosa/metabolismo , Homeostasis , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/genética , Obesidad/complicaciones , Ovariectomía , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Resultado del Tratamiento , Regulación hacia Arriba , Aumento de Peso
12.
Viruses ; 12(9)2020 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-32933138

RESUMEN

Repurposing FDA-approved compounds could provide the fastest route to alleviate the burden of disease caused by flaviviruses. In this study, three fluoroquinolones, enoxacin, difloxacin and ciprofloxacin, curtailed replication of flaviviruses Zika (ZIKV), dengue (DENV), Langat (LGTV) and Modoc (MODV) in HEK-293 cells at low micromolar concentrations. Time-of-addition assays suggested that enoxacin suppressed ZIKV replication at an intermediate step in the virus life cycle, whereas ciprofloxacin and difloxacin had a wider window of efficacy. A129 mice infected with 1 × 105 plaque-forming units (pfu) ZIKV FSS13025 (n = 20) or phosphate buffered saline (PBS) (n = 11) on day 0 and treated with enoxacin at 10 mg/kg or 15 mg/kg or diluent orally twice daily on days 1-5 did not differ in weight change or virus titer in serum or brain. However, mice treated with enoxacin showed a significant, five-fold decrease in ZIKV titer in testes relative to controls. Mice infected with 1 × 102 pfu ZIKV (n = 13) or PBS (n = 13) on day 0 and treated with 15 mg/kg oral enoxacin or diluent twice daily pre-treatment and days 1-5 post-treatment also did not differ in weight and viral load in the serum, brain, and liver, but mice treated with enoxacin showed a significant, 2.5-fold decrease in ZIKV titer in testes relative to controls. ZIKV can be sexually transmitted, so reduction of titer in the testes by enoxacin should be further investigated.


Asunto(s)
Antivirales/farmacología , Flavivirus/efectos de los fármacos , Fluoroquinolonas/farmacología , Replicación Viral/efectos de los fármacos , Animales , Ciprofloxacina/análogos & derivados , Ciprofloxacina/farmacología , Dengue , Virus del Dengue/efectos de los fármacos , Enoxacino/farmacología , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Testículo/virología , Carga Viral , Virus Zika/efectos de los fármacos
13.
Trends Pharmacol Sci ; 29(3): 116-23, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18262661

RESUMEN

Estrogen is a crucial hormone in human physiology that regulates a multitude of biological processes. It is also an important target in many diseases such as cancer and skeletal, neurological and immunological conditions. The actions of estrogen have traditionally been ascribed to one of two closely related classical nuclear hormone receptors, ERalpha and ERbeta, which are best characterized for regulating gene expression. Recent studies have revealed the contribution of a novel estrogen receptor GPR30, which belongs to the family of seven-transmembrane G-protein-coupled receptors, to many of the rapid biological responses to estrogen. Many drugs, such as tamoxifen and fulvestrant, which seem to selectively inhibit the activities of the classical estrogen receptors, are in widespread clinical use. However, recent results indicate that these same drugs activate multiple cellular-signaling pathways via GPR30. Unraveling the pharmacological profiles and specificities of ERalpha, ERbeta and GPR30 will be vital for understanding not only the physiological roles of each receptor but also for the development of the next generation of receptor-specific drugs.


Asunto(s)
Enfermedad , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Enfermedad/etiología , Congéneres del Estradiol/efectos adversos , Congéneres del Estradiol/farmacología , Congéneres del Estradiol/uso terapéutico , Humanos , Ligandos , Fitoestrógenos/efectos adversos , Fitoestrógenos/farmacología , Fitoestrógenos/uso terapéutico , Receptores de Estrógenos/genética , Receptores Acoplados a Proteínas G/genética
14.
J Neurosci Res ; 87(7): 1610-9, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19125412

RESUMEN

The G protein-coupled receptor GPR30 has recently been identified as a nonnuclear estrogen receptor. Reverse transcriptase-polymerase chain reaction revealed expression of GPR30 mRNA in varying quantities in the rat spinal cord, dorsal root ganglia, nodose ganglia, trigeminal ganglia, hippocampus, brain stem, and hypothalamus. Immunohistochemical studies that used a rabbit polyclonal antiserum against the human GPR30 C-terminus revealed a fine network of GPR30-immunoreactive (irGPR30) cell processes in the superficial layers of the spinal cord; some of which extended into deeper laminae. A population of neurons in the dorsal horn and ventral horn were irGPR30. Dorsal root, nodose, and trigeminal ganglionic neurons displayed varying intensities of irGPR30. Positively labeled neurons were detected in the major pelvic ganglion, but not in the superior cervical ganglion. A population of chromaffin cells in the adrenal medulla was irGPR30, so were cells of the zona glomerulosa. Double-labeling the adrenal medulla with GPR30 antiserum and tyrosine hydroxylase antibody or phenylethanolamine-N-methyltransferase antiserum revealed that irGPR30 is expressed in the majority of tyrosine hydroxylase-positive chromaffin cells. Last, some of the myenteric ganglion cells were irGPR30. Tissues processed with preimmune serum resulted in no staining. Voltage-sensitive dye imaging studies showed that the selective GPR30 agonist G-1 (1, 10, and 100 nM) depolarized cultured spinal neurons in a concentration-dependent manner. Collectively, our result provides the first evidence that GPR30 is expressed in neurons of the dorsal and ventral horn as well as in sensory and autonomic neurons, and activation of GPR30 by the selective agonist G-1 depolarizes cultured spinal neurons.


Asunto(s)
Ganglios Autónomos/metabolismo , Ganglios Sensoriales/metabolismo , Neuronas/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Médula Espinal/metabolismo , Animales , Tronco Encefálico/metabolismo , Células Cultivadas , Células Cromafines/metabolismo , Ciclopentanos/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Expresión Génica , Hipocampo/metabolismo , Hipotálamo/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Feniletanolamina N-Metiltransferasa/metabolismo , Quinolinas/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Tirosina 3-Monooxigenasa/metabolismo
15.
Cytometry A ; 75(3): 253-63, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18785269

RESUMEN

Of recent, clinical interest have been two related human G-protein coupled receptors: formylpeptide receptor (FPR), linked to antibacterial inflammation and malignant glioma cell metastasis; and FPR like-1 (FPRL1), linked to chronic inflammation in systemic amyloidosis, Alzheimer's disease, and prion diseases. In association with the National Institutes of Health (NIH) Molecular Library Screening Network, we implemented a flow-cytometry-based high-throughput screening (HTS) approach for identifying selective small molecule FPR and FPRL1 ligands. The screening assay measured the ability of test compounds to competitively displace a high-affinity, fluorescein- labeled peptide ligand from FPR, FPRL1, or both. U937 cells expressing FPR and rat basophil leukemia (RBL) cells expressing FPRL1 were tested together in a "duplex" format. The U937 cells were color coded with red-fluorescent dye allowing their distinction during analysis. Compounds, cells, and fluorescent ligand were sequentially combined (no wash) in 15 microl assay volumes in 384-well plates. Throughput averaged approximately 11 min per plate to analyze approximately 4,000 cells ( approximately 2,000/receptor) in a 2 microl aspirate from each well. In primary single concentration HTS of 24,304 NIH Small Molecule Repository compounds, 253 resulted in inhibition >30% (181 for FPR, 72 for FPRL1) of which 40 had selective binding inhibition constants (K(i)) < or = 4 microM (34 for FPR and 6 for FPRL1). An additional 1,446 candidate compounds were selected by structure-activity-relationship analysis of the hits and screened to identify novel ligands for FPR (3570-0208, K(i) = 95 +/- 10 nM) and FPRL1 (BB-V-115, K(i) = 270 +/- 51 nM). Each was a selective antagonist in calcium response assays and the most potent small molecule antagonist reported for its respective receptor to date. The duplex assay format reduced assay time, minimized reagent requirements, and provided selectivity information at every screening stage, thus proving to be an efficient means to screen for selective receptor ligand probes.


Asunto(s)
Citometría de Flujo/métodos , Receptores de Formil Péptido/antagonistas & inhibidores , Receptores de Lipoxina/antagonistas & inhibidores , Animales , Células Cultivadas , Factores Quimiotácticos/metabolismo , Colorantes Fluorescentes , Humanos , Ligandos , Sondas Moleculares/análisis , Ratas , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/metabolismo , Sensibilidad y Especificidad , Células U937
16.
Neuropharmacology ; 158: 107701, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31291595

RESUMEN

Multiple system atrophy (MSA) is a fatal demyelinating disorder lacking any disease-modifying therapies. MSA pathology stems from aggregated α-synuclein (aSyn) accumulation in glial cytosolic inclusions of oligodendroglial cell (OLGs), the myelinating cells of brain. In MSA brains and in MSA animal models with aSyn accumulation in OLGs, aberrant expression of brain-derived neurotrophic factor (BDNF) and glial-cell-line-derived neurotrophic factor (GDNF) occur. Nerve growth factor (NGF) expression can also be altered in neurodegenerative diseases. It is unclear if oxidative stress impacts the viability of aSyn-accumulating OLG cells. Here, we show that OLN-93 cells stably expressing human wild type aSyn or the MSA-associated-aSyn-mutants G51D or A53E, are more vulnerable to oxidative stress. In dose response studies we found that OLN-93 cells treated 48 h with 160 nM FTY720 or our new non-immunosuppressive FTY720-C2 or FTY720-Mitoxy derivatives sustained normal viability. Also, FTY720, FTY720-C2, and FTY720-Mitoxy all stimulated NGF expression at 24 h. However only FTY720-Mitoxy also increased BDNF and GDNF mRNA at 24 h, an effect paralleled by increases in histone 3 acetylation and ERK1/2 phosphorylation. Myelin associated glycoprotein (MAG) levels were also increased in OLN-93 cells after 48 h treatment with FTY720-Mitoxy. FTY720, FTY720-C2, and FTY720-Mitoxy all prevented oxidative-stress-associated-cell-death of OLN-93 cells that lack any aSyn expression. However, only FTY720-Mitoxy protected MSA-like aSyn-expressing-OLN-93-cells against oxidative-cell-death. These data identify potent protective effects for FTY720-Mitoxy with regard to trophic factors as well as MAG expression by OLG cells. Testing of FTY720-Mitoxy in mice is thus a judicious next step for neuropharmacological preclinical development.


Asunto(s)
Ceramidas/farmacología , Clorhidrato de Fingolimod/análogos & derivados , Atrofia de Múltiples Sistemas/metabolismo , Oligodendroglía/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Moduladores de los Receptores de fosfatos y esfingosina 1/farmacología , alfa-Sinucleína/efectos de los fármacos , Animales , Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Línea Celular , Clorhidrato de Fingolimod/farmacología , Factor Neurotrófico Derivado de la Línea Celular Glial/efectos de los fármacos , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Glicoproteína Asociada a Mielina/efectos de los fármacos , Glicoproteína Asociada a Mielina/metabolismo , Factor de Crecimiento Nervioso/efectos de los fármacos , Factor de Crecimiento Nervioso/metabolismo , Oligodendroglía/metabolismo , Ratas , alfa-Sinucleína/metabolismo
17.
ChemistryOpen ; 8(2): 201-205, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30815328

RESUMEN

Exquisite control of catalytic metathesis reactivity is possible through ligand-based variation of ruthenium carbene complexes. Sterically hindered alkenes, however, remain a generally recalcitrant class of substrates for intermolecular cross-metathesis. Allylic chalcogenides (sulfides and selenides) have emerged as "privileged" substrates that exhibit enhanced turnover rates with the commercially available second-generation ruthenium catalyst. Increased turnover rates are advantageous when competing catalyst degradation is limiting, although specific mechanisms have not been defined. Herein, we describe facile cross-metathesis of allylic sulfone reagents with sterically hindered isoprenoid alkene substrates. Furthermore, we demonstrate the first example of intermolecular cross-metathesis of ruthenium carbenes with a tetrasubstituted alkene. Computational analysis by combined coupled cluster/DFT calculations exposes a favorable energetic profile for metallacyclobutane formation from chelating ruthenium ß-chalcogenide carbene intermediates. These results establish allylic sulfones as privileged reagents for a substrate-based strategy of cross-metathesis derivatization.

18.
Cell Chem Biol ; 26(12): 1692-1702.e5, 2019 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-31706983

RESUMEN

Estrogen exerts extensive and diverse effects throughout the body of women. In addition to the classical nuclear estrogen receptors (ERα and ERß), the G protein-coupled estrogen receptor GPER is an important mediator of estrogen action. Existing ER-targeted therapeutic agents act as GPER agonists. Here, we report the identification of a small molecule, named AB-1, with the previously unidentified activity of high selectivity for binding classical ERs over GPER. AB-1 also possesses a unique functional activity profile as an agonist of transcriptional activity but an antagonist of rapid signaling through ERα. Our results define a class of small molecules that discriminate between the classical ERs and GPER, as well as between modes of signaling within the classical ERs. Such an activity profile, if developed into an ER antagonist, could represent an opportunity for the development of first-in-class nuclear hormone receptor-targeted therapeutics for breast cancer exhibiting reduced acquired and de novo resistance.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Ligandos , Transducción de Señal , Animales , Proliferación Celular/efectos de los fármacos , Estradiol/farmacología , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor beta de Estrógeno/antagonistas & inhibidores , Femenino , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Útero/efectos de los fármacos , Útero/metabolismo
19.
J Nucl Med ; 49(6): 978-86, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18483091

RESUMEN

UNLABELLED: Breast and endometrial cancers are the most common invasive malignancies in women, with more than 217,000 new diagnoses per year in the United States. These cancers are often classified into 2 subtypes based on the expression of the classical estrogen receptor. In this study, we describe a new structural class of neutral tridentate 99mTc(I)-estradiol-pyridin-2-yl hydrazine derivatives for potential use in breast and endometrial cancer imaging. METHODS: The 99mTc(I)-estradiol-pyridin-2-yl hydrazine derivative was synthesized via the Sonogashira cross-coupling reaction and radiolabeled via the tricarbonyl approach. Radiochemical purity was assessed by high-performance liquid chromatography. Cell-binding studies were performed with human breast adenocarcinoma MCF-7 cells. The in vivo biodistribution of the 99mTc(I) derivative was evaluated in virgin female C57BL/6 mice in defined phases of the estrous cycle. Biodistribution and SPECT/CT studies were performed with mice bearing MCF-7 and primary human endometrial tumors. RESULTS: Radiochemical analysis demonstrated that the postpurification purity of the 99mTc(I)-estradiol-pyridin-2-yl hydrazine derivative was > or =95%, with a specific activity of 99mTc of 47.5 TBq/mmol. Cell-binding studies yielded a dissociation constant (mean +/- SEM) of 11 +/- 1.5 nM. In vivo studies revealed that receptor-mediated uptake was present in all phases of the estrous cycle in reproductive organs and mammary glands but was highest during the diestrous phase of the estrous cycle. Despite high nonspecific uptake in the liver, significant receptor-mediated uptake was observed in target tissues and estrogen receptor-expressing tumors (0.67% for MCF-7 tumors and 0.77% for endometrial tumors). Tumor uptake was reduced by approximately 50% on coinjection with 17beta-estradiol. CONCLUSION: We have characterized a novel neutral tridentate 99mTc(I)-estradiol-pyridin-2-yl hydrazine derivative for potential use in breast and endometrial cancer imaging. This study represents the first step on a path toward the design of estrogen-based Tc-labeled tracers with improved targeting and SPECT imaging characteristics.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/metabolismo , Dihidrotestosterona/análogos & derivados , Neoplasias Endometriales/diagnóstico por imagen , Neoplasias Endometriales/metabolismo , Nandrolona/análogos & derivados , Receptores de Estrógenos/metabolismo , Animales , Línea Celular Tumoral , Dihidrotestosterona/farmacocinética , Sistemas de Liberación de Medicamentos/métodos , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Tasa de Depuración Metabólica , Ratones , Ratones Endogámicos C57BL , Nandrolona/farmacocinética , Especificidad de Órganos , Cintigrafía , Radiofármacos/farmacocinética , Distribución Tisular
20.
Antiviral Res ; 79(1): 19-27, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18394724

RESUMEN

There are no FDA approved drugs for the treatment of hemorrhagic fever with renal syndrome (HFRS), a serious human illnesses caused by hantaviruses. Clinical studies using ribavirin (RBV) to treat HFRS patients suggest that it provides an improved prognosis when given early in the course of disease. Given the unique antiviral activity of RBV and the lack of other lead scaffolds, we prepared a diverse series of 3-substituted 1,2,4-triazole-beta-ribosides and identified one with antiviral activity, 1-beta-d-ribofuranosyl-3-ethynyl-[1,2,4]triazole (ETAR). ETAR showed an EC(50) value of 10 and 4.4 microM for Hantaan virus (HTNV) and Andes virus, respectively. ETAR had weak activity against Crimean Congo hemorrhagic fever virus, but had no activity against Rift Valley fever virus. Intraperitoneally delivered ETAR offered protection to suckling mice challenged with HTNV with a approximately 25% survival at 12.5 and 25mg/kg ETAR, and a MTD of 17.1+/-0.7 days. ETAR was phosphorylated in Vero E6 cells to its 5'-triphosphate and reduced cellular GTP levels. In contrast to RBV, ETAR did not increase mutation frequency of the HTNV genome, which suggests it has a different mechanism of action than RBV. ETAR is an exciting and promising lead compound that will be elaborated in further synthetic investigations as a framework for the rational design of new antivirals for treatment of HFRS.


Asunto(s)
Antivirales/síntesis química , Antivirales/farmacología , Fiebre Hemorrágica con Síndrome Renal/tratamiento farmacológico , Nucleósidos/síntesis química , Nucleósidos/farmacología , Orthohantavirus/efectos de los fármacos , Triazoles/síntesis química , Triazoles/farmacología , Animales , Antivirales/metabolismo , Chlorocebus aethiops , Femenino , Genoma Viral/efectos de los fármacos , Guanosina/antagonistas & inhibidores , Guanosina/metabolismo , Guanosina Trifosfato/antagonistas & inhibidores , Guanosina Trifosfato/metabolismo , Orthohantavirus/genética , Orthohantavirus/metabolismo , Fiebre Hemorrágica con Síndrome Renal/virología , Humanos , Ratones , Ratones Endogámicos , Mutación/efectos de los fármacos , Nucleósidos/metabolismo , Ribavirina/análogos & derivados , Ribavirina/síntesis química , Ribavirina/metabolismo , Ribavirina/farmacología , Triazoles/metabolismo , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA