Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Med Chem ; 53(11): 4511-21, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20462211

RESUMEN

Structural modification of a virtual screening hit led to the identification of a new series of 4-[3-aryl-2,2-dioxido-2,1,3-benzothiadiazol-1(3H)-yl]-1-(methylamino)butan-2-ols which are potent and selective inhibitors of the norepinephrine transporter over both the serotonin and dopamine transporters. One representative compound S-17b (WYE-103231) had low nanomolar hNET potency (IC(50) = 1.2 nM) and excellent selectivity for hNET over hSERT (>1600-fold) and hDAT (>600-fold). S-17b additionally had a good pharmacokinetic profile and demonstrated oral efficacy in rat models of ovariectomized-induced thermoregulatory dysfunction and morphine dependent flush as well as the hot plate and spinal nerve ligation (SNL) models of acute and neuropathic pain.


Asunto(s)
Óxidos S-Cíclicos/química , Óxidos S-Cíclicos/farmacología , Descubrimiento de Drogas/métodos , Inhibidores de la Captación de Neurotransmisores/química , Inhibidores de la Captación de Neurotransmisores/farmacología , Norepinefrina/metabolismo , Tiadiazoles/química , Tiadiazoles/farmacología , Animales , Línea Celular , Óxidos S-Cíclicos/síntesis química , Óxidos S-Cíclicos/farmacocinética , Femenino , Humanos , Masculino , Inhibidores de la Captación de Neurotransmisores/síntesis química , Inhibidores de la Captación de Neurotransmisores/farmacocinética , Ratas , Relación Estructura-Actividad , Tiadiazoles/síntesis química , Tiadiazoles/farmacocinética
2.
Eur J Pharmacol ; 616(1-3): 334-9, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19576879

RESUMEN

Estrogens modulate critical homeostatic functions of the hypothalamus such as temperature regulation, sexual behavior and sleep with the most pronounced effects in rats occurring during the dark-phase. The neurochemical signals underlying estrogenic regulation of these hypothalamic functions have not been clearly identified, possibly due to the fact that previous studies have not explored the effects of estrogen treatments on neuronal signaling during the dark-phase. In the present study, ovariectomized rats received estradiol benzoate (5 microg/rat for 7 days, s.c.) and norepinephrine and dopamine levels were measured in the preoptic area of the hypothalamus across the light/dark cycle using in vivo microdialysis. Estradiol benzoate treatment increased extracellular norepinephrine and dopamine levels relative to vehicle treatment during the dark-phase. Increases in norepinephrine and dopamine were first detected by 30 min and 5.5h after lights-off, respectively. Subsequent increases in norepinephrine and dopamine were also noted throughout the 9.5-h collection period. The effect of estradiol benzoate on catecholamine release did not correlate with increases in either tyrosine hydroxylase (TH) protein expression or activity levels in the anterior hypothalamus, although a marked decrease in TH activity correlated with a rise in extracellular norepinephrine at the beginning of the dark-phase. We conclude that subchronic estradiol benzoate treatment increases extracellular catecholamine levels in the preoptic area of the hypothalamus during the dark-phase without a concomitant increase in neurotransmitter biosynthesis. The estradiol benzoate-induced increases in norepinephrine and dopamine levels in the preoptic area during the dark-phase may play an important role in modulating critical hypothalamic functions.


Asunto(s)
Catecolaminas/metabolismo , Oscuridad , Estradiol/análogos & derivados , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Ovariectomía , Animales , Dopamina/metabolismo , Estradiol/farmacología , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/metabolismo , Femenino , Hipotálamo/citología , Microdiálisis , Norepinefrina/metabolismo , Área Preóptica/citología , Área Preóptica/efectos de los fármacos , Área Preóptica/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Tirosina 3-Monooxigenasa/antagonistas & inhibidores
3.
J Neurosci Methods ; 182(2): 260-5, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19520113

RESUMEN

The Abeta1-42 fragment from the Amyloid Precursor Protein (APP) has presented considerable challenges from an analytical perspective. It is present at low levels in the circulation and can bind to proteins which mask its presence in assays. A number of therapeutic strategies target the lowering of this peptide, necessitating more robust and sensitive methods for its measurement. In this study, conditions for extracting and enriching Abeta1-42 using solid-phase extraction (SPE) and reverse-phase HPLC (RP-HPLC) were optimized. The new process provided reproducible recovery of Abeta1-42 of about 80% and allowed for concentration of the peptide prior to assay. Radiolabeled Abeta1-42 and ELISA for Abeta1-42 were used to determine the recovery and distribution of the peptide from whole blood collected in the presence of potassium-EDTA. Endogenous Abeta1-42 yielded a cell pellet:plasma ratio near 40:60 while exogenously added peptide distributed with a ratio of about 27:73. Additionally, the Abeta1-42 in the plasma and cell pellet fractions maintained stability over many hours. Comparing the measurement of Abeta1-42 using a commercial ELISA before and after enrichment demonstrated noticeable improvement of signal in samples enriched for the peptide. The current study also showed that conspicuous amounts of Abeta1-42 partition to the cell pellet but that this fraction can be robustly recovered and measured with SPE and HPLC. The process utilized established chromatographic techniques and is suitable for automation. It is also compatible with other detection methods including mass spectrometry.


Asunto(s)
Enfermedad de Alzheimer/sangre , Péptidos beta-Amiloides/sangre , Fragmentos de Péptidos/sangre , Cromatografía Líquida de Alta Presión , Ensayo de Inmunoadsorción Enzimática , Humanos , Masculino , Persona de Mediana Edad , Chaperonas Moleculares/metabolismo , Plasma/química , Unión Proteica , Extracción en Fase Sólida
4.
J Neurosci Methods ; 178(2): 270-5, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19135085

RESUMEN

Temperature dysfunction, clinically described as hot flashes/flushes and night sweats, commonly occur in women transitioning through menopause. Research in this field has yet to fully elucidate the biological underpinnings explaining this dysfunction. The need to develop animal models that can be used to study hormone-dependent temperature regulation is essential to advancing this scientific area. Development of telemetric transmitters for monitoring tail-skin (TST) and core body (CBT) temperatures for animal research has increased the accuracy of data by reducing extraneous factors associated with previous methods. However, until recently, TST and CBT could not be simultaneously measured telemetrically within the same animal. In this report, new dual temperature monitoring transmitters were validated by simultaneously evaluating them with the single measurement transmitters using the ovariectomized (OVX) rat thermoregulatory dysfunction model. A major advantage of measuring TST and CBT in the same animal is the ability to relate temporal changes on these two temperature parameters. Comparative experimentation was performed by single administration of clonidine (alpha(2) adrenergic agonist), MDL-100907 (5-HT(2a) antagonist), or a 7-day treatment of 17alpha-ethinyl estradiol (EE). Clonidine caused decreases in TST and CBT, MDL-100907 caused increases in TST while decreasing CBT, and EE caused decreases in TST with minor CBT decreases only at the higher dose. Data from either probe type showed similar results on temperature parameters regardless of transmitter used. These findings support the use of the new dual temperature transmitters and should enhance the quality and interpretation of data being generated in thermoregulation studies.


Asunto(s)
Regulación de la Temperatura Corporal , Temperatura Corporal , Monitoreo Fisiológico/métodos , Temperatura Cutánea , Telemetría/métodos , Agonistas alfa-Adrenérgicos/administración & dosificación , Animales , Temperatura Corporal/efectos de los fármacos , Clonidina/administración & dosificación , Modelos Animales de Enfermedad , Estradiol/administración & dosificación , Estrógenos/administración & dosificación , Femenino , Fluorobencenos/administración & dosificación , Ovariectomía , Piperidinas/administración & dosificación , Ratas , Ratas Sprague-Dawley , Antagonistas de la Serotonina/administración & dosificación , Temperatura Cutánea/efectos de los fármacos , Cola (estructura animal) , Factores de Tiempo
5.
Eur J Pharmacol ; 591(1-3): 315-8, 2008 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-18619956

RESUMEN

Estrogen treatment decreases active phase rapid eye movement (REM) sleep in ovariectomized rats. Here we explored further the effect of 17alpha-ethinyl estradiol (17alpha-EE) on active phase REM sleep in ovariectomized rats by analyzing spectral properties and the number and length of REM sleep bouts. The greatest suppression of REM sleep occurred on day 4 of 17alpha-EE treatment, was due to decreases in bout length, and was accompanied by decreased EEG theta power. These results further elucidate 17alpha-EE's effects on REM sleep and provide greater understanding of the mechanisms by which estrogens alter sleep-wakefulness patterns.


Asunto(s)
Estrógenos/farmacología , Etinilestradiol/farmacología , Sueño REM/efectos de los fármacos , Animales , Femenino , Ovariectomía , Polisomnografía , Ratas , Ratas Sprague-Dawley , Fases del Sueño/efectos de los fármacos , Ritmo Teta , Factores de Tiempo , Vigilia/efectos de los fármacos
6.
Endocrinology ; 149(10): 5219-26, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18599545

RESUMEN

Previous reports suggest the antiestrogen ICI 182,780 (ICI) does not cross the blood-brain barrier (BBB). However, this hypothesis has never been directly tested. In the present study, we tested whether ICI crosses the BBB, penetrates into brain and hypothalamic tissues, and affects known neuroendocrine functions in ovariectomized rats. Using HPLC with mass spectrometry, ICI (1.0 mg/kg.d, 3 d) was detected in plasma and brain and hypothalamic tissues for up to 24 h with maximum concentrations of 43.1 ng/ml, and 31.6 and 38.8 ng/g, respectively. To evaluate antiestrogenic effects of ICI in the brain after systemic dosing, we tested its ability to block the effect of 17 alpha-ethinyl estradiol (EE) (0.3 mg/kg, 8 d) on tail-skin temperature abatement in the morphine-dependent model of hot flush and on body weight change. In the morphine-dependent model, EE abated 64% of the naloxone-induced tail-skin temperature increase. ICI pretreatment (1.0, 3.0 mg/kg.d) dose dependently inhibited this effect. ICI (3.0 mg/kg.d) alone showed estrogenic-like actions, abating 30% the naloxone-induced flush. In body weight studies, EE-treated rats weighed 58.5 g less than vehicle-treated rats after 8 d dosing. This effect was partially blocked by ICI (3.0 mg/kg.d) pretreatment. Similar to EE treatment, rats receiving 1.0 or 3.0 mg/kg.d ICI alone showed little weight gain compared with vehicle-treated controls. Thus, ICI crosses the BBB, penetrates into brain and hypothalamic tissues, and has both antiestrogenic and estrogenic-like actions on neuroendocrine-related functions.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Estradiol/análogos & derivados , Antagonistas de Estrógenos/farmacocinética , Hipotálamo/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Estradiol/farmacocinética , Estradiol/farmacología , Antagonistas de Estrógenos/farmacología , Etinilestradiol/farmacología , Femenino , Fulvestrant , Sofocos/inducido químicamente , Sofocos/metabolismo , Morfina/farmacología , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Narcóticos/farmacología , Ovariectomía , Ratas , Ratas Sprague-Dawley , Temperatura Cutánea/efectos de los fármacos , Útero/efectos de los fármacos
7.
Horm Behav ; 53(1): 217-24, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17976600

RESUMEN

In ovariectomized (OVX) Sprague-Dawley rats, estradiol benzoate (EB) has been reported to decrease rapid eye movement (REM) and non-REM (NREM) sleep during the dark phase for up to 3 days. It is unknown, however, if estrogenic effects on sleep extend beyond 3 days or if other estrogens could induce the same changes. Furthermore, it is unclear whether the increased wakefulness in the dark phase was due to changes in active or quiet wakefulness. Therefore, we examined the effects of daily injections of 17alpha-ethinyl estradiol (EE) for 6 days on sleep and wakefulness in the OVX rat. After 3 days of baseline recording using a telemetric system, rats were administered sesame oil (sc) for 3 days followed by injection with EE (20 mug/rat/day, sc) for 6 days. After treatment, sleep was recorded during hormone withdrawal for an additional 5 days. A few sporadic but statistically significant increases in light phase sleep occurred during the last 3 days of EE treatment. Starting on day 2 of the study, EE caused statistically significant decreases in dark phase REM sleep that were maintained throughout the treatment period and persisted until the 3rd day of hormone withdrawal. During the dark phase, statistically significant decreases in NREM sleep and increases in active wakefulness started on the second day of treatment and abated by the end of treatment. This study demonstrated that EE had similar effects on sleep-wakefulness to EB and demonstrates the utility of telemetric polysomnographic recording of the female OVX rat as a model for understanding the estrogen-induced changes on sleep-wakefulness.


Asunto(s)
Etinilestradiol/administración & dosificación , Fases del Sueño/efectos de los fármacos , Vigilia/efectos de los fármacos , Animales , Vías de Administración de Medicamentos , Femenino , Ovariectomía , Ratas , Ratas Sprague-Dawley
8.
Endocrinology ; 148(3): 1376-83, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17122073

RESUMEN

Hot flushes and night sweats, referred to as vasomotor symptoms (VMS), are presumed to be a result of declining hormone levels and are the principal menopausal symptoms for which women seek medical treatment. To date, estrogens and/or some progestins are the most effective therapeutics for alleviating VMS; however, these therapies may not be appropriate for all women. Therefore, nonhormonal therapies are being evaluated. The present study investigated a new reuptake inhibitor, desvenlafaxine succinate (DVS), in animal models of temperature dysfunction. Both models used are based on measuring changes in tail-skin temperature (TST) in ovariectomized (OVX) rats. The first relies on naloxone-induced withdrawal in morphine-dependent (MD) OVX rats, resulting in an acute rise in TST. The second depends on an OVX-induced loss of TST decreases during the dark phase as measured by telemetry. An initial evaluation demonstrated abatement of the rise in TST with long-term administration of ethinyl estradiol or with a single oral dose of DVS (130 mg/kg) in the MD model. Further evaluation showed that orally administered DVS acutely and dose dependently (10-100 mg/kg) abated a naloxone-induced rise in TST of MD rats and alleviated OVX-induced temperature dysfunction in the telemetry model. Oral administration of DVS to OVX rats caused significant increases in serotonin and norepinephrine levels in the preoptic area of the hypothalamus, a key region of the brain involved in temperature regulation. These preclinical studies provide evidence that DVS directly impacts thermoregulatory dysfunction in OVX rats and may have utility in alleviating VMS associated with menopause.


Asunto(s)
Regulación de la Temperatura Corporal/efectos de los fármacos , Ciclohexanoles/farmacología , Ovariectomía , Administración Oral , Antagonistas Adrenérgicos alfa/farmacología , Animales , Ciclohexanoles/administración & dosificación , Succinato de Desvenlafaxina , Evaluación Preclínica de Medicamentos , Etinilestradiol/farmacología , Femenino , Modelos Animales , Dependencia de Morfina/patología , Norepinefrina/antagonistas & inhibidores , Área Preóptica/química , Área Preóptica/efectos de los fármacos , Ratas , Antagonistas de la Serotonina/farmacología , Telemetría
9.
Neuroendocrinology ; 84(5): 330-8, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17192701

RESUMEN

Thermoregulation is an integrated network of neuroendocrine, autonomic and somatosensory responses. Thermoregulatory dysfunction occurs during fluctuations or decline of gonadal hormone levels and results in vasomotor symptoms such as hot flushes and/or night-time sweating. The neurotransmitter serotonin (5-HT), has been reported to play a role in thermoregulation via changes in extracellular 5-HT levels and/or activation of various 5-HT receptors. The purpose of this study was to evaluate the role of the selective 5-HT reuptake inhibitor (SSRI), fluoxetine (FLX), on temperature regulation using ovariectomized (OVX) rodent models of thermoregulation. Single, subcutaneous (s.c.) administration of FLX (3, 10, 30 and 60 mg/kg) dose-dependently reduced core body temperature (CBT). FLX at 3 and 10 mg/kg s.c. showed no statistically significant decrease on tail-skin temperature (TST), whereas at higher doses (30 and 60 mg/kg) a significant decrease in TST was noted in the telemetry model. To mimic chronic SSRI treatment, a 5-HT(1A) antagonist (WAY-100635; 0.3 mg/kg) was administered 20 min prior to FLX (10 mg/kg). This combination showed no significant improvement on temperature dysfunction compared to FLX alone. Similarly, in a morphine-dependent model of temperature dysfunction FLX, was inactive at 10 mg/kg whereas the 30 and 60 mg/kg s.c. dose abated the naloxone-induced increase in TST by 55 and 81%, respectively. In summary, FLX affected CBT at all doses, but alleviated thermoregulatory dysfunction only at higher doses that are non-selective for the 5-HT system.


Asunto(s)
Regulación de la Temperatura Corporal/efectos de los fármacos , Fluoxetina/farmacología , Ovariectomía , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Femenino , Fluoxetina/administración & dosificación , Sofocos/patología , Sofocos/terapia , Dependencia de Morfina/patología , Naloxona/farmacología , Ratas , Ratas Sprague-Dawley , Antagonistas del Receptor de Serotonina 5-HT1 , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Telemetría
10.
Brain Res ; 1123(1): 135-44, 2006 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-17067560

RESUMEN

Thermoregulation is a complex intercommunicative function requiring coordination between core body temperature (CBT), the central nervous system, and peripheral vasculature. In menopausal women, dysregulation of thermoregulatory mechanisms leads to hot flushes and night sweats. A previous study in ovariectomized (OVX) rats has suggested that mirtazapine can alleviate thermoregulatory dysfunction by blocking 5-HT(2A) receptor signaling. This is in opposition to other work in which 5-HT(2A) receptor blockade appeared to exacerbate thermoregulatory dysfunction in OVX rats. Thus, the goals of the present study were to reexamine the effects of mirtazapine on temperature regulation in OVX rat models and explore further the role of 5-HT(2A) receptor blockade. Mirtazapine exhibited potent functional antagonism (EC(50)=0.62 nM) at the cloned human 5-HT(2A) receptor. In the morphine-dependent model of thermoregulatory dysfunction, mirtazapine (10 mg/kg, i.p.) induced an increase in tail-skin temperature (TST) prior to naloxone administration. In the telemetry model, mirtazapine (0.3-3 mg/kg, i.p.) caused an increase in TST. However, at the highest dose tested (10 mg/kg, i.p.), mirtazapine induced a small but significant decrease in TST followed by an increase in TST. To examine this finding further, mirtazapine's effect on CBT was determined. Administration of mirtazapine (1-3 mg/kg, i.p.) resulted in a slight decrease in CBT but at the 10 mg/kg dose a dramatic decrease (-3.6 degrees C) in CBT was observed. These data support the concept that 5-HT(2A) receptors play a role in temperature regulation but that functional blockade of these receptors by mirtazapine is not a likely mechanism for restoring thermoregulatory processes in OVX rats.


Asunto(s)
Regulación de la Temperatura Corporal/efectos de los fármacos , Sofocos/prevención & control , Mianserina/análogos & derivados , Antagonistas del Receptor de Serotonina 5-HT2 , Antagonistas de la Serotonina/farmacología , Animales , Relación Dosis-Respuesta a Droga , Femenino , Mianserina/farmacología , Mirtazapina , Ovariectomía , Ratas , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT2A/metabolismo
11.
Brain Res ; 1098(1): 71-8, 2006 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-16764833

RESUMEN

Desvenlafaxine succinate (DVS) is a novel serotonin (5-HT) and norepinephrine (NE) reuptake inhibitor (SNRI) that is currently in clinical development for the treatment of major depressive disorder and vasomotor symptoms associated with menopause. Previous studies have documented the pharmacokinetic and pharmacodynamic profiles of DVS in male rats. Similar studies, however, have not been performed in ovariectomized (OVX) rats, a model that mimics the loss of ovarian hormones that occurs at menopause. The goal of the present study, therefore, was to characterize the pharmacokinetic and pharmacodynamic properties of DVS in OVX rats. Desvenlafaxine levels peaked in plasma, brain (total brain minus hypothalamus) and hypothalamus at concentrations of 7.0, 10.8 and 9.5 microM (assuming 1 g = 1 ml), respectively, 30 min post-dosing DVS (30 mg/kg, oral). The apparent terminal half-lives of desvenlafaxine in plasma, brain and hypothalamus were 3.0, 2.1 and 2.5 h, respectively. Based on AUC(0-last), brain to plasma and hypothalamus to plasma ratios were 1.7 and 1.3, respectively. Microdialysis experiments in the medial preoptic area of the hypothalamus showed that DVS (30 mg/kg, s.c.), in the presence of WAY-100635 (5-HT(1A) antagonist), increased 5-HT levels 225% at 1 h post-dosing. Norepinephrine levels increased 44% at 3 h post-dosing while dopamine levels were unchanged. Thus, in OVX rats, DVS has good pharmacokinetic properties, rapid brain penetration, excellent brain penetrability and selectively increases 5-HT and NE levels in the hypothalamus. This work supports the notion that DVS could have utility for treating disorders in menopausal women in which changes in 5-HT and/or NE have been implicated.


Asunto(s)
Inhibidores de Captación Adrenérgica/farmacología , Inhibidores de Captación Adrenérgica/farmacocinética , Ciclohexanoles/farmacología , Ciclohexanoles/farmacocinética , Ovariectomía , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacocinética , Animales , Química Encefálica/efectos de los fármacos , Succinato de Desvenlafaxina , Dopamina/metabolismo , Femenino , Microdiálisis , Norepinefrina/metabolismo , Área Preóptica/efectos de los fármacos , Área Preóptica/metabolismo , Ratas , Ratas Sprague-Dawley , Serotonina/metabolismo , Técnicas Estereotáxicas
12.
J Comp Neurol ; 496(6): 773-86, 2006 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-16628620

RESUMEN

The identity and biology of stem cells and progenitors in the adult brain are of considerable interest, because these cells hold great promise for the development of novel therapies for damaged brain tissue in human diseases. This research field critically needs biological markers that specifically identify the resident precursors in the germinal zones of the adult central nervous system so that the discovery of regulatory influences for adult neurogenesis may be facilitated. In this study, by using a combination of in situ hybridization, bromodeoxyuridine incorporation, immunocolocalization, and ultrastructural studies, we show that in rodents Tctex-1, a cytoplasmic dynein light chain, is selectively enriched in almost all cycling progenitors and young neuronal progeny, but not in mature granular cells and astrocytes, in the subgranular zone of the adult dentate gyrus. Tctex-1 is also selectively abundant in cells closely resembling previously described immature progenitors and migrating neuroblasts at the subventricular zone of the lateral ventricle. Our results suggest that Tctex-1 serves as a novel marker for the identification of neural progenitors of the adult brain.


Asunto(s)
Astrocitos/metabolismo , Giro Dentado/citología , Giro Dentado/metabolismo , Proteínas Asociadas a Microtúbulos/biosíntesis , Neuronas/metabolismo , Proteínas Nucleares/biosíntesis , Células Madre/metabolismo , Animales , Biomarcadores/análisis , Bromodesoxiuridina , Giro Dentado/ultraestructura , Proteínas de Dominio Doblecortina , Dineínas , Femenino , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/biosíntesis , Neuropéptidos/biosíntesis , Proteínas de Unión al ARN/biosíntesis , Ratas , Ratas Sprague-Dawley , Región del Complejo T del Genoma
13.
J Neurosci ; 23(34): 10982-7, 2003 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-14645494

RESUMEN

Mitochondrial dysfunction may contribute to dopaminergic (DAergic) cell death in Parkinson's disease and GABAergic cell death in Huntington's disease. In the present work, we tested whether blocking A1 receptors could enhance the damage to DAergic and GABAergic neurons caused by mitochondrial inhibition, and whether blocking A2a receptors could protect against damage in this model. Animals received an intraperitoneal injection of 8-cyclopentyl-1,3-dipropylxanthine (CPX) (A1 antagonist) or 3,7-dimethyl-1-propargylxanthine (DMPX) (A2a antagonist) 30 min before intrastriatal infusion of malonate (mitochondrial complex II inhibitor). Damage was assessed 1 week later by measuring striatal dopamine, tyrosine hydroxylase (TH), and GABA content. In mice and rats, malonate-induced depletion of striatal dopamine, TH, or GABA was potentiated by pretreatment with 1 mg/kg CPX and attenuated by pretreatment with 5 mg/kg DMPX. To determine the location of the A1 and A2a receptors mediating these effects, CPX or DMPX was infused directly into the striatum or substantia nigra of rats 30 min before intrastriatal infusion of malonate. When infused into the striatum, CPX (20 ng) potentiated, whereas DMPX (50 ng) prevented malonate-induced GABA loss, but up to 100 ng of CPX or 500 ng of DMPX did not alter malonate-induced striatal dopamine loss. Intranigral infusion of CPX (100 ng) or DMPX (500 ng), however, did exacerbate and protect, respectively, against malonate-induced striatal dopamine loss. Thus, A1 receptor blockade enhances and A2a receptor blockade protects against damage to DAergic and GABAergic neurons caused by mitochondrial inhibition. Interestingly, these effects are mediated by A1 and A2a receptors located in the substantia nigra for DAergic neurons and in the striatum for GABAergic neurons.


Asunto(s)
Adenosina/metabolismo , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Mitocondrias/metabolismo , Sustancia Negra/metabolismo , Teobromina/análogos & derivados , Ácido gamma-Aminobutírico/metabolismo , Antagonistas del Receptor de Adenosina A1 , Antagonistas del Receptor de Adenosina A2 , Animales , Cuerpo Estriado/efectos de los fármacos , Vías de Administración de Medicamentos , Sinergismo Farmacológico , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Enfermedad de Huntington/metabolismo , Masculino , Malonatos/farmacología , Ratones , Mitocondrias/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Trastornos Parkinsonianos/metabolismo , Ratas , Ratas Sprague-Dawley , Sustancia Negra/efectos de los fármacos , Teobromina/farmacología , Tirosina 3-Monooxigenasa/metabolismo , Xantinas/farmacología
14.
J Neurochem ; 83(3): 665-72, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12390528

RESUMEN

The neuromodulator adenosine mediates dark-adaptive changes in retinal photoreceptors through A(2a) receptors. In cold-blooded vertebrates, opsin mRNA expression is lower at night than during the day. In the present study, we tested whether adenosine could inhibit opsin mRNA expression in cultured rod cells and if endogenous adenosine acts to suppress opsin mRNA in the intact retina at night. Semi-quantitative in situ hybridization showed that treatment with 100 nm of the A(2a)/A(2b) agonist N(6)-[2-(3,5-dimethoxyphenyl)-2-(2-methylphenyl)-ethyl]adenosine (DPMA) reduced opsin mRNA 41% in cultured rod cells. The effect of DPMA was blocked by 10 microm of the A(2a) antagonist 8-(3-chlorostyryl)caffeine (CSC) but not by 10 microm of the A(2b) antagonist alloxazine. One micromolar adenosine alone had no effect on opsin mRNA. However, in the presence of the adenosine deaminase inhibitor erythro-9-(2-hydroxy-3-nonyl)adenine hydrochloride (EHNA), 1 microm adenosine reduced opsin mRNA 61%. EHNA alone reduced opsin mRNA by 26%. Consistent with an A(2a) receptor mechanism, 100 nm forskolin (adenylate cyclase agonist) decreased opsin mRNA 34%. Finally, northern blots showed that intravitreal injection of 10 microm CSC at night increased opsin I mRNA 38%. Thus, endogenous adenosine suppresses rod opsin I mRNA expression at night; in vitro results indicate this reduction occurs through A(2a)-like receptor binding and stimulation of adenylate cyclase activity.


Asunto(s)
Adenina/análogos & derivados , Adenosina/análogos & derivados , Regulación de la Expresión Génica/fisiología , ARN Mensajero/biosíntesis , Receptores Purinérgicos P1/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Opsinas de Bastones/biosíntesis , Adenina/farmacología , Adenosina/farmacología , Inhibidores de la Adenosina Desaminasa , Adenilil Ciclasas/efectos de los fármacos , Adenilil Ciclasas/metabolismo , Ambystoma , Animales , Células Cultivadas , Colforsina/farmacología , Oscuridad , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Agonistas del Receptor Purinérgico P1 , ARN Mensajero/antagonistas & inhibidores , Receptor de Adenosina A2A , Retina/citología , Retina/efectos de los fármacos , Retina/metabolismo , Células Fotorreceptoras Retinianas Bastones/citología , Células Fotorreceptoras Retinianas Bastones/efectos de los fármacos , Opsinas de Bastones/genética
15.
Proc Natl Acad Sci U S A ; 99(8): 5655-60, 2002 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-11943854

RESUMEN

Rod photoreceptors are highly compartmentalized sensory neurons that maintain strict ultrastructural and molecular polarity. Structural subdivisions include the outer segment, inner segment, cell body, and synaptic terminal. The visual pigment rhodopsin is found predominantly in membranes of the rod cell outer segment but becomes mislocalized, appearing throughout the plasma membrane of the cell in many retinal diseases and injuries. Currently, there is no known link between rhodopsin redistribution and rod cell death. We propose that activation of mislocalized rhodopsin kills rod cells by stimulating normally inaccessible signaling pathways. This hypothesis was tested in primary retinal cell cultures, which contain photoreceptors. In rod photoreceptors, opsin immunofluorescence occurred throughout the rod cell plasma membrane. Activation of this mislocalized opsin by photostimulation after formation of isorhodopsin or by incubation with beta-ionone (opsin agonist) killed 19-30% of rod cells. Rod cell death was apoptotic, as indicated by marked chromatin condensation and the requirement for caspase-3 activation. Rod cell death could be induced by forskolin (adenylate cyclase agonist), and conversely, beta-ionone-induced cell death could be blocked by cotreatment with SQ22536 (an adenylate cyclase inhibitor). Pertussis toxin (a G protein inhibitor) also blocked beta-ionone-induced cell death. The data support a mechanism by which activation of mislocalized opsin initiates apoptotic rod cell death through G protein stimulation of adenylate cyclase.


Asunto(s)
Adenina/análogos & derivados , Enfermedades de la Retina/metabolismo , Células Fotorreceptoras Retinianas Bastones/efectos de los fármacos , Células Fotorreceptoras Retinianas Bastones/patología , Opsinas de Bastones/farmacología , Adenina/farmacología , Toxina de Adenilato Ciclasa , Adenilil Ciclasas/metabolismo , Animales , Apoptosis , Caspasa 3 , Caspasas/metabolismo , Muerte Celular , Células Cultivadas , Cromatina/metabolismo , Colforsina/metabolismo , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Microscopía Fluorescente , Modelos Biológicos , Toxina del Pertussis , Transducción de Señal , Factores de Tiempo , Urodelos , Factores de Virulencia de Bordetella/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...