Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Pharmacol Exp Ther ; 348(3): 372-82, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24338505

RESUMEN

AS1069562 [(R)-2-[(1H-inden-7-yloxy)methyl]morpholine monobenzenesulfonate] is the (+)-isomer of indeloxazine, which had been used clinically for the treatment of cerebrovascular diseases with multiple pharmacological actions, including serotonin (5-HT) and norepinephrine (NE) reuptake inhibition. Here we investigated the analgesic effects of AS1069562 in a rat model of chronic constriction injury (CCI)-induced neuropathic pain and the spinal monoamine turnover. These effects were compared with those of the antidepressants duloxetine and amitriptyline. AS1069562 significantly elevated extracellular 5-HT and NE levels in the rat spinal dorsal horn, although its 5-HT and NE reuptake inhibition was much weaker than that of duloxetine in vitro. In addition, AS1069562 increased the ratio of the contents of both 5-HT and NE to their metabolites in rat spinal cord, whereas duloxetine slightly increased only the ratio of the content of 5-HT to its metabolite. In CCI rats, AS1069562 and duloxetine significantly ameliorated mechanical allodynia, whereas amitriptyline did not. AS1069562 and amitriptyline significantly ameliorated thermal hyperalgesia, and duloxetine tended to ameliorate it. Furthermore, AS1069562, duloxetine, and amitriptyline significantly improved spontaneous pain-associated behavior. In a gastric emptying study, AS1069562 affected gastric emptying at the same dose that exerted analgesia in CCI rats. On the other hand, duloxetine and amitriptyline significantly reduced gastric emptying at lower doses than those that exerted analgesic effects. These results indicate that AS1069562 broadly improved various types of neuropathic pain-related behavior in CCI rats with unique characteristics in spinal monoamine turnover, suggesting that AS1069562 may have potential as a treatment option for patients with neuropathic pain, with a different profile from currently available antidepressants.


Asunto(s)
Analgésicos/uso terapéutico , Monoaminas Biogénicas/metabolismo , Morfolinas/uso terapéutico , Neuralgia/tratamiento farmacológico , Médula Espinal/efectos de los fármacos , Amitriptilina/farmacología , Analgésicos/farmacocinética , Analgésicos/farmacología , Animales , Dopamina/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Clorhidrato de Duloxetina , Vaciamiento Gástrico/efectos de los fármacos , Células HEK293 , Humanos , Masculino , Morfolinas/farmacocinética , Morfolinas/farmacología , Actividad Motora/efectos de los fármacos , Neuralgia/fisiopatología , Norepinefrina/metabolismo , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Unión Proteica , Ratas , Ratas Sprague-Dawley , Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Médula Espinal/metabolismo , Estereoisomerismo , Tiofenos/farmacocinética , Tiofenos/farmacología
2.
J Neurosci ; 32(6): 2037-50, 2012 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-22323718

RESUMEN

γ-Secretase inhibitors (GSIs) reduce amyloid-ß (Aß) peptides but inevitably increase the ß-C-terminal fragment (ß-CTF) of amyloid precursor protein (APP), potentially having undesirable effects on synapses. In contrast, γ-secretase modulators (GSMs) reduce Aß42 without increasing ß-CTF. Although the Aß-lowering effects of these compounds have been extensively studied, little effort has been made to investigate their effects on cognition. Here, we compared the effects of two GSIs--(2S)-2-hydroxy-3-methyl-N-[(2S)-1-{[(1S)-3-methyl-2-oxo-2,3,4,5-tetrahydro-1H-3-benzazepin-1-yl]amino}-1-oxopropan-2-yl]butanamide (LY450139, semagacestat) and (2R)-2-[[(4-chlorophenyl)sulfonyl][[2-fluoro-4-(1,2,4-oxazol-3-yl)phenyl]methyl]amino-5,5,5-trifluoropentanamide (BMS-708163)--and a second-generation GSM [{(2S,4R)-1-[(4R)-1,1,1-trifluoro-7-methyloctan-4-yl]-2-[4-(trifluoromethyl)phenyl]piperidin-4-yl}acetic acid (GSM-2)] on spatial working memory in APP-transgenic (Tg2576) and nontransgenic mice using the Y-maze task. While acute dosing with either GSI ameliorated memory deficits in 5.5-month-old Tg2576 mice, these effects disappeared after 8 d subchronic dosing. Subchronic dosing with either GSI rather impaired normal cognition in 3-month-old Tg2576 mice, with no inhibition on the processing of other γ-secretase substrates, such as Notch, N-cadherin, or EphA4, in the brain. LY450139 also impaired normal cognition in wild-type mice; however, the potency was 10-fold lower than that in Tg2576 mice, indicating an APP-dependent mechanism likely with ß-CTF accumulation. Immunofluorescence studies revealed that the ß-CTF accumulation was localized in the presynaptic terminals of the hippocampal stratum lucidum and dentate hilus, implying an effect on presynaptic function in the mossy fibers. In contrast, both acute and subchronic dosing with GSM-2 significantly ameliorated memory deficits in Tg2576 mice and did not affect normal cognition in wild-type mice. We demonstrated a clear difference between GSI and GSM in effects on functional consequences, providing new insights into strategies for developing these drugs against Alzheimer's disease.


Asunto(s)
Alanina/análogos & derivados , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/fisiología , Precursor de Proteína beta-Amiloide/fisiología , Azepinas/farmacología , Cognición/efectos de los fármacos , Cognición/fisiología , Inhibidores de Proteasas/farmacología , Alanina/farmacología , Precursor de Proteína beta-Amiloide/genética , Animales , Línea Celular Tumoral , Femenino , Humanos , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos
3.
J Neurochem ; 125(3): 465-72, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23240999

RESUMEN

Given that amyloid-ß 42 (Aß42) is believed to be a culprit in Alzheimer's disease (AD), reducing Aß42 production should be a potential therapeutic approach. γ-Secretase modulators (GSMs) cause selective reduction of Aß42 or both reduction of Aß42 and Aß40 without affecting total Aß through shifting the γ-cleavage position in amyloid precursor protein. We recently reported on GSM-2, one of the second-generation GSMs, that selectively reduced brain Aß42 level and significantly ameliorated cognitive deficits in plaque-free 5.5-month-old Tg2576 AD model mice. Here, we investigated the effects of GSM-2 on 10-, 14-, and 18-month-old mice which had age-dependent increase in amyloid plaques. Eight-day treatment with GSM-2 significantly ameliorated cognitive deficits measured by Y-maze task in the mice of any age. However, GSM-2 reduced brain soluble Aß42 only in 10-month-old mice. In contrast, GSM-2 markedly reduced newly synthesized soluble Aß42 in both 10- and 18-month-old mice with similar efficacy when measured using the stable isotope-labeling technique, suggesting that nascent Aß42 plays a more significant role than plaque-associated soluble Aß42 in the cognitive deterioration of Tg2576 mice. These findings further indicate the potential utility of approach to reducing Aß42 synthesis in AD therapeutic regimens.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Trastornos del Conocimiento/metabolismo , Inhibidores Enzimáticos/uso terapéutico , Fragmentos de Péptidos/metabolismo , Acetatos/farmacología , Acetatos/uso terapéutico , Factores de Edad , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Péptidos beta-Amiloides/inmunología , Precursor de Proteína beta-Amiloide/genética , Animales , Anticuerpos/uso terapéutico , Cromatografía Liquida , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/etiología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/química , Ensayo de Inmunoadsorción Enzimática , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Espectrometría de Masas , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Fragmentos de Péptidos/inmunología , Piperidinas/farmacología , Piperidinas/uso terapéutico
4.
J Anesth ; 27(6): 939-41, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23760511

RESUMEN

Voltage-dependent sodium channels (VDSCs) are crucial for pain generation. Here, to develop a new behavioral index of pain induced by spinal VDSC activation, we examined whether intrathecal veratridine injection produced nociceptive behavior. Intrathecal injection of the VDSC opener veratridine in mice dose-dependently induced nociceptive responses, with response times subsequently reduced by administration of morphine or pregabalin. Systemic administration of lidocaine and mexiletine, but not amitriptyline, also decreased this response time. Taken together, these results demonstrated that response time of nociceptive behavior induced by intrathecal veratridine injection is a quantitative index of pain triggered by spinal VDSC activation.


Asunto(s)
Dolor Nociceptivo/inducido químicamente , Manejo del Dolor/métodos , Dimensión del Dolor/métodos , Columna Vertebral/efectos de los fármacos , Agonistas del Canal de Sodio Activado por Voltaje/farmacología , Canales de Sodio Activados por Voltaje/metabolismo , Amitriptilina/farmacología , Animales , Inyecciones Espinales , Lidocaína/farmacología , Masculino , Mexiletine/farmacología , Ratones , Ratones Endogámicos ICR , Morfina/farmacología , Dolor Nociceptivo/tratamiento farmacológico , Pregabalina , Columna Vertebral/metabolismo , Ácido gamma-Aminobutírico/análogos & derivados , Ácido gamma-Aminobutírico/farmacología
5.
J Pharmacol Exp Ther ; 336(3): 743-50, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21098091

RESUMEN

Transient receptor potential vanilloid 1 (TRPV1) is activated by a variety of stimulations, such as endogenous ligands and low pH, and is believed to play a role in pain transmission. TRPV1 antagonists have been reported to be effective in several animal pain models; however, some compounds induce hyperthermia in animals and humans. We discovered the novel TRPV1 antagonist (R)-N-(1-methyl-2-oxo-1,2,3,4-tetrahydro-7-quinolyl)-2-[(2-methylpyrrolidin-1-yl)methyl]biphenyl-4-carboxamide (AS1928370) in our laboratory. AS1928370 bound to the resiniferatoxin-binding site on TRPV1 and inhibited capsaicin-mediated inward currents with an IC50 value of 32.5 nM. Although AS1928370 inhibited the capsaicin-induced Ca²(+) flux in human and rat TRPV1-expressing cells, the inhibitory effect on proton-induced Ca²(+) flux was extremely small. In addition, AS1928370 showed no inhibitory effects on transient receptor potential vanilloid 4, transient receptor potential ankyrin 1, and transient receptor potential melastatin 8 in concentrations up to 10 µM. AS1928370 improved capsaicin-induced secondary hyperalgesia and mechanical allodynia in an L5/L6 spinal nerve ligation model in rats with respective ED50 values of 0.17 and 0.26 mg/kg p.o. Furthermore, AS1928370 alleviated inflammatory pain in a complete Freund's adjuvant model at 10 mg/kg p.o. AS1928370 had no effect on rectal body temperature up to 10 mg/kg p.o., although a significant hypothermic effect was noted at 30 mg/kg p.o. In addition, AS1928370 showed no significant effect on motor coordination. These results suggest that blockage of the TRPV1 receptor without affecting the proton-mediated TRPV1 activation is a promising approach to treating neuropathic pain because of the potential wide safety margin against hyperthermic effects. As such, compounds such as ASP1928370 may have potential as new analgesic agents for treating neuropathic pain.


Asunto(s)
Analgésicos/uso terapéutico , Benzamidas/uso terapéutico , Fiebre , Neuralgia/tratamiento farmacológico , Dimensión del Dolor/efectos de los fármacos , Quinolonas/uso terapéutico , Canales Catiónicos TRPV/antagonistas & inhibidores , Analgésicos/farmacología , Animales , Benzamidas/química , Benzamidas/farmacología , Capsaicina/farmacología , Capsaicina/uso terapéutico , Fiebre/inducido químicamente , Células HEK293 , Humanos , Masculino , Neuralgia/fisiopatología , Dolor/tratamiento farmacológico , Dolor/fisiopatología , Dimensión del Dolor/métodos , Unión Proteica/fisiología , Quinolonas/química , Quinolonas/farmacología , Ratas , Ratas Sprague-Dawley , Canales Catiónicos TRPV/fisiología
6.
Biol Pharm Bull ; 34(7): 1105-8, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21720020

RESUMEN

Transient receptor potential vanilloid 1 (TRPV1) is primarily expressed in central and peripheral terminals of non-myelinated primary afferent neurons. We previously showed that AS1928370, a novel TRPV1 antagonist that can prevent ligand-induced activation but not proton-induced activation, ameliorates neuropathic pain in rats without hyperthermic effect. In this study, we investigated its analgesic profile in mice. AS1928370 showed good oral bioavailability and high penetration into the brain and spinal cord in mice. The mean plasma-to-brain and plasma-to-spinal cord ratios were 4.3 and 3.5, respectively. Pretreatment with AS1928370 significantly suppressed both capsaicin-induced acute pain and withdrawal response in hot plate test at 10-30 mg/kg per os (p.o.). At lower oral doses (0.3-1.0 mg/kg), AS1928370 improved mechanical allodynia in mice undergoing spinal nerve ligation. Intrathecal administration of AS1928370 (30 µg/body) also significantly suppressed mechanical allodynia. In addition, AS1928370 showed no effect on locomotor activity up to 30 mg/kg p.o. These results suggest that spinal TRPV1 has an important role in the transmission of neuropathic pain and that the central nervous system (CNS) penetrant TRPV1 receptor antagonist AS1928370 is a promising candidate for treating neuropathic pain.


Asunto(s)
Benzamidas/farmacología , Hiperalgesia/prevención & control , Neuralgia/prevención & control , Quinolonas/farmacología , Canales Catiónicos TRPV/antagonistas & inhibidores , Animales , Benzamidas/administración & dosificación , Benzamidas/farmacocinética , Capsaicina/farmacología , Modelos Animales de Enfermedad , Inyecciones Espinales , Ratones , Neuralgia/inducido químicamente , Quinolonas/administración & dosificación , Quinolonas/farmacocinética , Distribución Tisular
7.
Proc Natl Acad Sci U S A ; 105(16): 6133-8, 2008 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-18413613

RESUMEN

The G protein-coupled receptor (GPCR) family is highly diversified and involved in many forms of information processing. SREB2 (GPR85) is the most conserved GPCR throughout vertebrate evolution and is expressed abundantly in brain structures exhibiting high levels of plasticity, e.g., the hippocampal dentate gyrus. Here, we show that SREB2 is involved in determining brain size, modulating diverse behaviors, and potentially in vulnerability to schizophrenia. Mild overexpression of SREB2 caused significant brain weight reduction and ventricular enlargement in transgenic (Tg) mice as well as behavioral abnormalities mirroring psychiatric disorders, e.g., decreased social interaction, abnormal sensorimotor gating, and impaired memory. SREB2 KO mice showed a reciprocal phenotype, a significant increase in brain weight accompanying a trend toward enhanced memory without apparent other behavioral abnormalities. In both Tg and KO mice, no gross malformation of brain structures was observed. Because of phenotypic overlap between SREB2 Tg mice and schizophrenia, we sought a possible link between the two. Minor alleles of two SREB2 SNPs, located in intron 2 and in the 3' UTR, were overtransmitted to schizophrenia patients in a family-based sample and showed an allele load association with reduced hippocampal gray matter volume in patients. Our data implicate SREB2 as a potential risk factor for psychiatric disorders and its pathway as a target for psychiatric therapy.


Asunto(s)
Encéfalo/patología , Predisposición Genética a la Enfermedad/genética , Proteínas del Tejido Nervioso/genética , Receptores Acoplados a Proteínas G/genética , Esquizofrenia/genética , Esquizofrenia/patología , Alelos , Secuencia de Aminoácidos , Animales , Conducta Animal , Evolución Molecular , Humanos , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Tamaño de los Órganos/genética , Polimorfismo de Nucleótido Simple , Psicología del Esquizofrénico
8.
PLoS One ; 16(4): e0250663, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33905439

RESUMEN

In a disease-state-dependent manner, the histamine-resistant itch in dry skin-based skin diseases such as atopic dermatitis (AD) and xerosis is mainly due to hyperinnervation in the epidermis. Semaphorin 3A (Sema3A) is a nerve repulsion factor expressed in keratinocytes and it suppresses nerve fiber elongation in the epidermis. Our previous studies have shown that Sema3A ointment inhibits epidermal hyperinnervation and scratching behavior and improves dermatitis scores in AD model mice. Therefore, we consider Sema3A as a key therapeutic target for improving histamine-resistant itch in AD and xerosis. This study was designed to screen a library of herbal plant extracts to discover compounds with potential to induce Sema3A in normal human epidermal keratinocytes (NHEKs) using a reporter gene assay, so that positive samples were found. Among the positive samples, only the extract of S. baicalensis was found to consistently increase Sema3A levels in cultured NHEKs in assays using quantitative real-time PCR and ELISA. In evaluation of reconstituted human epidermis models, the level of Sema3A protein in culture supernatants significantly increased by application of the extract of S. baicalensis. In addition, we investigated which components in the extract of S. baicalensis contributed to Sema3A induction and found that baicalin and baicalein markedly increased the relative luciferase activity, and that baicalein had higher induction activity than baicalin. Thus, these findings suggest that S. baicalensis extract and its compounds, baicalin and baicalein, may be promising candidates for improving histamine-resistant itch via the induction of Sema3A expression in epidermal keratinocytes.


Asunto(s)
Extractos Vegetales/química , Scutellaria baicalensis/química , Semaforina-3A/metabolismo , Línea Celular , Flavanonas/genética , Flavanonas/metabolismo , Flavonoides/genética , Flavonoides/metabolismo , Genes Reporteros , Humanos , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Modelos Biológicos , Extractos Vegetales/farmacología , ARN Mensajero/metabolismo , Scutellaria baicalensis/metabolismo , Semaforina-3A/genética
9.
Biol Pharm Bull ; 33(1): 67-71, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20045938

RESUMEN

Dorsal root ganglia (DRG) are recognized as one of the organs which are damaged in peripheral sensory diabetic neuropathy. In an experimental animal model, the alteration of the mRNA expression level of neurotrophins, their receptors and neuronal cytoskeletal protein have been reported. In this study, we examined whether these changes are improved by treatment with the aldose reductase inhibitor, zenarestat, in early-stage diabetic neuropathy of streptozotocin (STZ)-induced diabetic rats. Two weeks after the induction of diabetes mellitus by STZ treatment, zenarestat or a vehicle were given orally for two weeks. After the zenarestat treatment, the mRNA expression levels of neurotrophin receptors and neuronal cytoskeletal proteins in dorsal root ganglia were determined with a real-time polymerase chain reaction (PCR) method. Compared with the expression level of normal rats, a significant increase in Trk-C and Talpha1 alpha-tubulin and a decrease in neurofilament H mRNA expression level were observed in the DRG of STZ rats, while there were no significant changes in Trk-A, Trk-B, p75, neurofilament L, neurofilament M and betaIII tubulin mRNA expression. Zenarestat treatment significantly ameliorated the abnormal increase in Trk-C mRNA expression level. These data suggest that hyperactivation of the polyol pathway induces a deficit in neurotropism on peripheral sensory diabetic neuropathy.


Asunto(s)
Aldehído Reductasa/antagonistas & inhibidores , Proteínas del Citoesqueleto/metabolismo , Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/metabolismo , Ganglios Espinales/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Animales , Proteínas del Citoesqueleto/genética , Inhibidores Enzimáticos/farmacología , Ganglios Espinales/efectos de los fármacos , Masculino , Proteínas del Tejido Nervioso/genética , Proteínas de Neurofilamentos/genética , Proteínas de Neurofilamentos/metabolismo , Neuronas/metabolismo , Polímeros/metabolismo , Quinazolinas/farmacología , ARN Mensajero/metabolismo , Ratas , Receptores de Factor de Crecimiento Nervioso/efectos de los fármacos , Receptores de Factor de Crecimiento Nervioso/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
10.
J Med Food ; 22(3): 257-263, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30543483

RESUMEN

Compound K (CK) is a metabolite of a saponin in Panax ginseng, formed from ginsenoside, a triterpenoid glycoside, by human intestinal bacteria. Lactobacillus paracasei A221 isolated from fermented food can hydrolyze (deglycosylate) the main ginsenoside, ginsenoside Rb1, and generate CK. However, the pharmacokinetics of L. paracasei A221 fermented ginseng (FG) and nonfermented ginseng (NFG) have not been investigated so far. The aim of this study was to investigate the pharmacokinetics of CK after oral administration of single doses of FG and NFG in healthy Japanese adults. An open-label, randomized, single-dose, two-period, crossover study was conducted in 12 Japanese healthy volunteers (five men and seven women, aged 40-60 years). All subjects were equally allocated into two groups and administered tablets containing FG or NFG. Until 24 h after the administration, blood samples were sequentially collected, plasma concentrations of CK were measured, and the pharmacokinetic parameters were calculated. We also expected restoration of decreased testosterone level as one of the beneficial effects of FG and measured plasma total testosterone concentrations in male volunteers. The means of Tmax, Cmax, and area under the concentration-time curve (AUC) were significantly different between the two groups. In the FG group, AUC0-12h (ng h/mL) and AUC0-24h (ng h/mL) were, respectively, 58.3- and 17.5-fold higher than those in the NFG group. Moreover, mean testosterone concentration in the FG group significantly increased 24 h after administration. These results showed that the main ginsenoside metabolite of ginseng, CK, produced by L. paracasei A221 has potential utility in health maintenance in healthy middle-aged and old Japanese adults.


Asunto(s)
Ginsenósidos/farmacocinética , Lacticaseibacillus paracasei/metabolismo , Panax/microbiología , Adulto , Estudios Cruzados , Femenino , Fermentación , Ginsenósidos/administración & dosificación , Ginsenósidos/sangre , Humanos , Japón , Masculino , Persona de Mediana Edad , Panax/química , Testosterona/sangre
11.
Pharmacol Ther ; 115(2): 292-306, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17599430

RESUMEN

Nerve growth factor (NGF) and other members of the neurotrophin family are critical for the survival and differentiation of neurons and have been implicated in the pathophysiology of numerous disease states. Although the therapeutic potential of neurotrophins has generated much excitement over the past decade, inconvenient pharmacokinetics and adverse side-effect profiles have limited the clinical usefulness of neurotrophic factors themselves. Compounds that mimic neurotrophin signaling and overcome the pharmacokinetic and side-effect barriers may have greater therapeutic potential. Here, we review the progress to date of clinical trials with direct neurotrophin modulators and describe alternative strategies to target (modulate) neurotrophin production and/or their signal transduction pathways. Particular emphasis is placed on small molecules that are able to modulate neurotrophin function in diseases of the nervous system. These alternative strategies show promise in preclinical studies, with some advancing into clinical development. Moreover, the recognition that clinically effective therapeutics, such as antidepressants and immunophilin ligands, can modulate neurotrophin function suggests that the concept of small molecule therapeutics that promote neurotrophic function may still be viable.


Asunto(s)
Factores de Crecimiento Nervioso/fisiología , Receptores de Factor de Crecimiento Nervioso/efectos de los fármacos , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Factor Neurotrófico Derivado del Encéfalo/fisiología , Factor Neurotrófico Derivado del Encéfalo/uso terapéutico , Depresión/tratamiento farmacológico , Depresión/metabolismo , Humanos , Factor de Crecimiento Nervioso/biosíntesis , Factor de Crecimiento Nervioso/fisiología , Factor de Crecimiento Nervioso/uso terapéutico , Factores de Crecimiento Nervioso/biosíntesis , Factores de Crecimiento Nervioso/uso terapéutico , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Enfermedades del Sistema Nervioso Periférico/metabolismo , Receptores de Factor de Crecimiento Nervioso/fisiología , Transducción de Señal , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/metabolismo , Cicatrización de Heridas/efectos de los fármacos
12.
Neuropharmacology ; 55(7): 1226-30, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18760290

RESUMEN

FK1706, a derivative of FK506, is a non-immunosuppressive immunophilin ligand with significant neurotrophic activity mediated via FKBP-52 and the RAS/RAF/MAPK signaling pathway. Here, we tested the effect of FK1706 on painful diabetic neuropathy in rat model of diabetes induced by streptozotocin (STZ). FK1706 ameliorated mechanical allodynia in this model at doses over 0.32 mg/kg, p.o., even if treatment was initiated after neuropathy was established, and did not affect plasma glucose levels. Furthermore, this improvement continued at least 4 weeks after the last administration. In morphological analysis, FK1706 treatment also restored intraepidermal nerve fiber density in footpad skin to almost normal levels. Gabapentin also improved mechanical allodynia in the same model, but efficacy disappeared the day after administration stopped. Allodynia responses were potentiated by co-administration of both compounds. Thus, FK1706 ameliorated painful diabetic neuropathy via a different mechanism from gabapentin and improved morphological outcomes, indicating that FK1706 improves painful diabetic neuropathy by modifying the underlying disease pathology.


Asunto(s)
Neuropatías Diabéticas/tratamiento farmacológico , Inmunofilinas/química , Dolor/tratamiento farmacológico , Tacrolimus/análogos & derivados , Aminas/farmacología , Analgésicos/farmacología , Animales , Ácidos Ciclohexanocarboxílicos/farmacología , Diabetes Mellitus Experimental/complicaciones , Neuropatías Diabéticas/psicología , Relación Dosis-Respuesta a Droga , Gabapentina , Inmunohistoquímica , Ligandos , Masculino , Dolor/etiología , Dimensión del Dolor/efectos de los fármacos , Estimulación Física , Ratas , Ratas Sprague-Dawley , Tacrolimus/farmacología , Ácido gamma-Aminobutírico/farmacología
13.
J Nucl Med ; 49(7): 1183-8, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18552135

RESUMEN

UNLABELLED: The purpose of the present study was to measure adenosine A(2A) receptor (A(2A)R) occupancy in the brain by a novel adenosine A(1)/A(2A) antagonist, 5-[5-amino-3-(4fluorophenyl)pyrazin-2-yl]-1-isopropylpyridine-2(1H)-one (ASP5854), and to determine the degree of receptor occupancy necessary to inhibit haloperidol-induced catalepsy in rhesus monkeys. METHODS: A(2A)R occupancy by ASP5854 (0.001-0.1 mg/kg) was examined in the striatum using an A(2A)R-specific radiotracer, (11)C-SCH442416, and PET in conscious rhesus monkeys. A(2A)R occupancy was monitored after a single intravenous administration of ASP5854 in 3 animals, and a dynamic PET scan was performed at 1, 4, and 8 h after an intravenous bolus injection of the tracer for approximately 740 MBq. Catalepsy was induced by haloperidol (0.03 mg/kg, intramuscularly) and examined for incidence and duration. RESULTS: ASP5854 dose-dependently increased A(2A)R occupancy in the striatum and showed long-lasting occupancy even after the reduction of plasma concentration. Haloperidol induced severe catalepsy at 40 min after intramuscular injection. The incidence and duration of cataleptic posture were dose-dependently reduced by ASP5854 at 1 h after oral administration, and the minimum ED(50) value was 0.1 mg/kg. Administration of a dose of 0.1 mg/kg yielded a plasma concentration of 97 +/- 16.3 ng/mL, which corresponded to 85%-90% of A(2A)R occupancy. CONCLUSION: These results showed that ASP5854 antagonized A(2A)R in the striatum, and the dissociation from A(2A)R was relatively slow. In addition, more than 85% A(2A)R occupancy by ASP5854 resulted in an inhibition of haloperidol-induced catalepsy. Thus, such a pharmacodynamic study directly demonstrates both the kinetics of a drug in the brain and the relationship between dose-dependent receptor occupancy and plasma level.


Asunto(s)
Antagonistas del Receptor de Adenosina A1 , Aminopiridinas/metabolismo , Cuerpo Estriado/metabolismo , Piperazinas/metabolismo , Receptor de Adenosina A2A/metabolismo , Antagonistas del Receptor de Adenosina A2 , Aminopiridinas/uso terapéutico , Animales , Radioisótopos de Carbono , Catalepsia/inducido químicamente , Catalepsia/tratamiento farmacológico , Cuerpo Estriado/diagnóstico por imagen , Haloperidol , Macaca mulatta , Masculino , Piperazinas/uso terapéutico , Tomografía de Emisión de Positrones , Pirazoles/metabolismo , Pirimidinas/metabolismo
14.
Behav Brain Res ; 194(2): 152-61, 2008 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-18657577

RESUMEN

Recent evidence indicates that adenosine A(2A) receptor antagonists hold therapeutic potential for the treatment of Parkinson's disease (PD). A study on the novel adenosine A(1) and A(2A) receptor dual antagonist 5-[5-amino-3-(4-fluorophenyl)pyrazin-2-yl]-1-isopropylpyridine-2(1H)-one (ASP5854) showed it to be effective in various rodents models of PD and cognition. In the present study, we further investigated the potential of ASP5854 as an anti-PD drug using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated common marmosets, which is a highly predictive model of clinical efficacy in PD, and compared its effect with those of existing anti-PD drugs. ASP5854 significantly and dose-dependently improved the total motor disability score for 7h at doses higher than 1mg/kg, and significantly increased total locomotor activity at doses higher than 0.1mg/kg without adverse effects. l-3,4-Dihydroxyphenylalanine+benserazide and bromocriptine also significantly improved the motor disability score and the hypolocomotion caused by MPTP treatment in a dose-dependent fashion. This amelioration was significant at 32+8 and 10-32 mg/kg, respectively, although bromocriptine induced severe emesis. Trihexiphenidyl also significantly improved the total motor disability score at doses of 10-32 mg/kg; however, while a significant increase in the total locomotor activity was observed at 10mg/kg, the drug induced ataxia-like behavior at 32 mg/kg. On the other hand, neither selegiline nor amantadine improved the total motor disability and hypolocomotion. These data substantiate the evidence that the novel adenosine antagonist ASP5854 exerts comparable anti-PD activity with existing anti-PD drugs, which indicates that ASP5854 might have potential to ameliorate motor deficits in PD.


Asunto(s)
Aminopiridinas/uso terapéutico , Antiparkinsonianos/uso terapéutico , Intoxicación por MPTP/complicaciones , Trastornos del Movimiento/tratamiento farmacológico , Trastornos del Movimiento/etiología , Piperazinas/uso terapéutico , Antagonistas Purinérgicos , Análisis de Varianza , Animales , Callithrix , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Intoxicación por MPTP/inducido químicamente , Masculino , Actividad Motora/efectos de los fármacos , Factores de Tiempo
15.
Eur J Pharmacol ; 591(1-3): 147-52, 2008 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-18602914

RESUMEN

Injured spinal cord axons fail to regenerate in part due to a lack of trophic support. While various methods for replacing neurotrophins have been pursued, clinical uses of these methods face significant barriers. FK1706, a non-immunosuppressant neurophilin ligand, potentiates nerve growth factor signaling, suggesting therapeutic potential for functional deficits following spinal cord injury. Here, we demonstrate that FK1706 significantly improves behavioral outcomes in animal models of spinal cord hemisection and contusion injuries in rats. Furthermore, we show that FK1706 is effective even if administration is delayed until 1 week after injury, suggesting that FK1706 has a reasonable therapeutic time-window. Morphological analysis of injured axons in the dorsal corticospinal tract showed an increase in the radius and perimeter of stained axons, which were reduced by FK1706 treatment, suggesting that axonal swelling and retraction balls observed in injured spinal cord were improved by the neurotrophic effect of FK1706. Taken together, FK1706 improves both behavioral motor function and the underlying morphological changes, suggesting that FK1706 may have therapeutic potential in meeting the significant unmet needs in spinal cord injury.


Asunto(s)
Regeneración Nerviosa/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Traumatismos de la Médula Espinal/tratamiento farmacológico , Tacrolimus/análogos & derivados , Animales , Axones/efectos de los fármacos , Axones/metabolismo , Modelos Animales de Enfermedad , Inmunofilinas/farmacología , Masculino , Factor de Crecimiento Nervioso/efectos de los fármacos , Factor de Crecimiento Nervioso/metabolismo , Tractos Piramidales/metabolismo , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Traumatismos de la Médula Espinal/fisiopatología , Tacrolimus/farmacología , Factores de Tiempo
16.
Pharmacol Biochem Behav ; 89(1): 11-6, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18067955

RESUMEN

We have recently confirmed that exposure of rats to the single-prolonged stress (SPS) paradigm induces enhanced hypothalamic-pituitary-adrenal (HPA) axis negative feedback and enhanced anxiety, and found that these changes develop time-dependently following stress exposure, suggesting that it could model the neuroendocrinological and behavioral abnormalities of the post-traumatic stress disorder (PTSD) patients. In the present study, microarray analysis was performed using RNA from the hippocampus, amygdala and anterior cingulate cortex of SPS rats and unstressed controls to unveil the molecular changes underlying SPS-induced behavioral changes. Thirty-one genes were found whose time course of expression corresponded to that of behavioral changes. One gene, 5-hydroxytryptamine2C (5-HT2C) receptor, was identified as a putative candidate. The overexpression of the gene in the amygdala of SPS rats was confirmed using real-time PCR 7 days after the SPS exposure. This molecule was then pharmacologically validated using FR260010 (N-[3-(4-methyl-1H-imidazol-1-yl)phenyl]-5,6-dihydrobenzo[h]quinazolin-4-amine dimethanesulfonate), a selective 5-HT2C receptor antagonist. FR260010 (1-10 mg/kg, s.c.) significantly inhibited the enhancement of anxiety in SPS rats. These results demonstrate for the first time that activation of the brain 5-HT2C receptor is involved in the development of behavioral abnormality in this model. This suggests that selective 5-HT2C receptor antagonists might provide novel therapeutic avenues for PTSD treatment.


Asunto(s)
Ansiedad/psicología , Receptor de Serotonina 5-HT2C/fisiología , Estrés Psicológico/psicología , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Ansiedad/etiología , Ansiedad/genética , Condicionamiento Psicológico/fisiología , Miedo/fisiología , Giro del Cíngulo/efectos de los fármacos , Giro del Cíngulo/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN/biosíntesis , ARN/genética , Ratas , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT2C/efectos de los fármacos , Receptor de Serotonina 5-HT2C/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Antagonistas de la Serotonina/farmacología , Estrés Psicológico/complicaciones , Estrés Psicológico/genética
17.
J Pharm Sci ; 107(11): 2883-2890, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30055224

RESUMEN

Heparinoid is commonly used for the treatment of superficial thrombophlebitis, a condition wherein inflammation and clotting occurs in the veins below the skin surface. However, stratum corneum is a major barrier that limits the delivery of hydrophilic heparinoid, in and across the skin. The aim of the present study was to develop a nonirritant topical formulation for heparinoid incorporating chemical penetration enhancers and investigate the delivery of heparinoid across the human epidermis using in vitro vertical Franz diffusion cells. The developed oil-in-water nanoemulsions (NEs; NE-1 and NE-2) delivered higher amount of heparinoid (91.58 ± 25.75 µg/sq.cm and 62.67 ± 5.66 µg/sq.cm, respectively) after 72 h compared with the other developed formulations, which in turn also delivered significantly higher amount compared with commercial formulations: cream (1.78 ± 0.07 µg/sq.cm), ointment (9.95 ± 4.41 µg/sq.cm), and gel (0 µg/sq.cm) (p <0.05). Transmission electron microscopy, polarizing light microscopy, and dynamic light scattering studies were performed to characterize the microstructure of these NEs with chemical enhancers. NE-1 was tested to be nonirritant with cell viability greater than 50% and a minimal release of IL-1α by using the "in vitro Epiderm tissue" model. Our results demonstrate that NE formulations represent a potential strategy for providing a localized therapy for the treatment of superficial thrombophlebitis.


Asunto(s)
Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacocinética , Epidermis/metabolismo , Heparinoides/administración & dosificación , Heparinoides/farmacocinética , Vehículos Farmacéuticos/química , Absorción Cutánea , Administración Cutánea , Composición de Medicamentos , Emulsiones/química , Humanos , Permeabilidad , Solubilidad , Termodinámica
18.
Brain Res ; 1149: 181-90, 2007 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-17391653

RESUMEN

The immunosuppressant cyclosporin A (CsA) has been shown to exert potent neuroprotective effects, possibly via the inhibition of calcineurin and mitochondrial permeability transition pore formation. Here, we investigated the neuroprotective profile of a novel derivative of CsA, FR901459, by evaluating its effects against in vitro mitochondrial damage and in vivo brain damage in transient global or focal cerebral ischemia models, in comparison with those of CsA. Efficacy of calcineurin inhibition was estimated from its immunosuppressive effect on the mixed lymphocyte reaction. Results showed that the immunosuppressive effect of FR901459 was approximately 7-fold less potent than that of CsA. In contrast, FR901459 suppressed Ca(2+)-induced mitochondrial swelling measured in isolated liver mitochondria with greater potency than CsA. Further, FR901459 showed approximately 30-fold greater neuroprotective potency than CsA against neuronal cell damage induced by thapsigargin in SH-SY5Y cells. In a transient global cerebral ischemia model in gerbils, FR901459 showed the dose-dependent suppression of neuronal cell death, while FR901459 was less efficacious than CsA. In a rat transient focal ischemia model, FR901459 tended to reduce brain damage on both intravenous injection as well as intracerebroventricular infusion, but with less efficacy than CsA which significantly reduced the damage. These findings suggest that FR901459 exerts a potent neuroprotective effect by inhibiting mitochondrial damage in vitro, but that in in vivo transient cerebral ischemia, its immunosuppressive component which possibly acts via the inhibition of calcineurin may play a more important role in attenuating brain damage than its inhibitory effect against mitochondrial damage.


Asunto(s)
Ciclosporina/farmacología , Hipoxia Encefálica/prevención & control , Ataque Isquémico Transitorio/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Animales , Línea Celular Tumoral , Gerbillinae , Humanos , Hipoxia Encefálica/etiología , Técnicas In Vitro , Ataque Isquémico Transitorio/complicaciones , Prueba de Cultivo Mixto de Linfocitos , Mitocondrias/patología , Ratas
19.
Eur J Pharmacol ; 563(1-3): 40-8, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17343843

RESUMEN

Mitochondrial membrane potential plays an important role in cell survival. Transitions in mitochondrial permeability, which indicate the imminent destruction of the organelles, have been observed in damaged neuronal cells both in vitro and in vivo. In this study, C57/BL6n mouse thymocytes were put under stress using thapsigargin, a Ca2+ ATP-ase inhibitor, after which the change in mitochondrial membrane potential was monitored with a JC-1 dual-emission probe. This was done in an attempt to identify a novel compound that can suppress mitochondrial membrane potential reduction and cell death. In this assay system, the novel compound SCH-20148 [2,3-dihydroxypropyl-5-bromo-N-(2-methyl-3-trifluoromethylphenyl)anthranilate] was found to protect mouse thymocytes against thapsigargin (3 nM)-induced mitochondrial membrane potential reduction (IC50=42 nM). SCH-20148 also prevented A23187- or ionomycin-induced shifts in mitochondrial membrane potential but it did not have any effect on the changes induced by tunicamycin, staurosporine, or dexamethasone. The potent immunosuppressants tacrolimus and cyclosporine A prevented the effect of thapsigargin, but did not prevent the A23187- or ionomycin-induced changes. Calcium-modulating agents, an anti-oxidant, a protein kinase C inhibitor, and anti-inflammatory agents were not effective against thapsigargin-induced mitochondrial permeability transition which implies that SCH-20148 exerts a protective effect via its specific mechanism. In addition, SH-20148 demonstrated a neuroprotective effect against thapsigargin-induced neuronal cell death in neuroblastoma SH-SY5Y cells. Taken together, these results suggest the potential of SCH-20148 as novel neuroprotective drug.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Membranas Mitocondriales/efectos de los fármacos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Tapsigargina/farmacología , Timo/efectos de los fármacos , ortoaminobenzoatos/farmacología , Animales , Calcimicina/farmacología , Calcio/metabolismo , ATPasas Transportadoras de Calcio/antagonistas & inhibidores , ATPasas Transportadoras de Calcio/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ciclosporina/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Inmunosupresores/farmacología , Ionomicina/farmacología , Ionóforos/farmacología , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Neuronas/metabolismo , Neuronas/patología , Permeabilidad/efectos de los fármacos , Tacrolimus/farmacología , Timo/metabolismo , Timo/patología
20.
J Biomol Screen ; 11(2): 155-64, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16361696

RESUMEN

Measurement of neurite outgrowth is a common assay of neurotrophic activity. However, currently available techniques for measuring neurite outgrowth are either time or resource intensive. The authors established a system in which chronic treatment of a subcloned SH-SY5Y cell line with aphidicolin and various concentrations of nerve growth factor (NGF) induced discernable alterations in proliferation and differentiation. Cells were fixed, labeled with a nonfluorescent dye, and evaluated both manually and with an automated analysis system. NGF increased multiple parameters of differentiation, including neurite length, the proportion of cells extending neurites, and branching, as well as promoting cellular survival/proliferation. Interestingly, although NGF treatment increased the total number of branches, it actually decreased the proportion of branches per neurite length. The authors observed no differences in results obtained using the manual and automated systems, but the automated system was orders of magnitude faster. To demonstrate the flexibility of the system, the authors also show that they could measure changes in differentiation induced by a small-molecule Rho kinase inhibitor, as well as by retinoic acid cotreatment with brain-derived neurotrophic factor. In addition to this flexibility, this system does not require specialized equipment or fluorescent antibodies for analysis and therefore provides a less resource-intensive alternative to fluorescence-based systems.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/farmacología , Factor de Crecimiento Nervioso/farmacología , Neuritas/química , Tretinoina/farmacología , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Afidicolina/farmacología , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Clonales , Sinergismo Farmacológico , Humanos , Neuritas/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA