Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Pharmacol Exp Ther ; 369(3): 364-374, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30918068

RESUMEN

Disease modification in Parkinson's disease (PD) is an unmet medical need. In the current study, we evaluated trehalose, a safe and well-tolerated disaccharide that has previously demonstrated efficacy in rodent models of neurodegenerative diseases, including PD. In a rat model of PD, based on delivery of adeno-associated virus serotype 1/2 containing the mutated human A53T α-synuclein gene (AAV1/2-hourA53T-aSyn) to the substantia nigra (SN), we showed that rats administered trehalose (2.67 g/kg per day, by mouth) for 6 weeks had less forelimb asymmetry (93% reduction) and higher striatal dopamine (54% increase) compared with rats receiving vehicle. In a pharmacokinetic study, we determined that efficacy was associated with plasma C max of 8900 ng/ml and area under the curve from time 0 to infinity (AUC0-inf) of 11,136 hour⋅ng/ml. We then showed, in macaques, that oral administration of trehalose (2.67 g/kg per day) produced plasma exposures of similar magnitude, with plasma C max of 10,918 ng/ml and AUC0-inf of 27,445 hour⋅ng/ml. In a macaque model of PD, also based on delivery of AAV1/2-hourA53T-aSyn to the SN, trehalose (2.67 g/kg per day, by mouth), administered for 142 days, produced higher striatal dopamine (by 39%) and dopamine transporter levels (by 50%), compared with macaques receiving vehicle. In neither model did trehalose treatment prevent loss of tyrosine hydroxylase (TH) positive (TH+ve) cells in the SN or alter α-synuclein levels in the striatum. These studies demonstrated that trehalose reduces striatal dopaminergic deficits in a rodent and macaque model of synucleinopathy in PD. Furthermore, we have determined the pharmacokinetic parameters associated with efficacy, and thus defined exposures to target in future clinical trials.


Asunto(s)
Dopamina/metabolismo , Neostriado/efectos de los fármacos , Neostriado/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Trehalosa/farmacología , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Macaca fascicularis , Enfermedad de Parkinson/sangre , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Ratas , Distribución Tisular , Trehalosa/sangre , Trehalosa/farmacocinética , Trehalosa/uso terapéutico
2.
J Pharmacol Exp Ther ; 367(2): 373-381, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30087157

RESUMEN

Dyskinesia is a common motor complication associated with the use of levodopa to treat Parkinson's disease. Numerous animal studies in mice, rats, and nonhuman primates have demonstrated that the N-methyl-d-aspartate antagonist, amantadine, dose dependently reduces levodopa-induced dyskinesia (LID). However, none of these studies characterized the amantadine plasma concentrations required for a therapeutic effect. This study evaluates the pharmacokinetic (PK)/pharmacodynamic (PD) relationship between amantadine plasma concentrations and antidyskinetic efficacy across multiple species to define optimal therapeutic dosing. The PK profile of amantadine was determined in mice, rats, and macaques. Efficacy data from the 6-hydroxydopamine rat and the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine macaque model of LID, along with previously published antidyskinetic efficacy data, were used to establish species-specific PK/PD relationships using a direct-effect maximum possible effect model. Results from the PK/PD model were compared with amantadine plasma concentrations and antidyskinetic effect in a phase 2 study in patients with Parkinson's disease treated with ADS-5102, an extended-release amantadine capsule formulation. Outcomes from each of the species evaluated indicate that the EC50 of amantadine for reducing dyskinesia range from 1025 to 1633 ng/ml (1367 ng/ml for an all-species model). These data are consistent with the mean amantadine plasma concentrations observed in patients with Parkinson's disease (∼1500 ng/ml) treated with ADS-5102 at doses that demonstrated a statistically significant reduction in dyskinesia. These results demonstrate that the EC50 of amantadine for reducing dyskinesia is consistent across multiple species and supports a plasma concentration target of ∼1400 ng/ml to achieve therapeutic efficacy.


Asunto(s)
Amantadina/farmacología , Amantadina/farmacocinética , Discinesia Inducida por Medicamentos/tratamiento farmacológico , Levodopa/farmacología , Animales , Antiparkinsonianos/farmacocinética , Antiparkinsonianos/farmacología , Modelos Animales de Enfermedad , Discinesia Inducida por Medicamentos/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Ratas , Ratas Sprague-Dawley
3.
FASEB J ; 22(7): 2488-97, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18364399

RESUMEN

Many experimental data support the enhancement of neurotrophic factors as a means to modify neurodegeneration in Parkinson's disease. However, the translation of this to the clinic has proven problematic. This is likely due to the complex nature of the surgical gene delivery and cell-based approaches adopted to deliver proteinaceous neurotrophic factors to targets within the central nervous system. We investigated the ability of a novel, orally active, nonpeptide neurotrophic factor inducer, PYM50028 (Cogane), to restore dopaminergic function after 1-methyl-4-phenylpyridinium (MPP(+)) -induced damage to mesencephalic neurons in vitro and in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) -lesioned mice. In rat mesencephalic neurons, administration of PYM50028, either before or after MPP(+), significantly prevented and reversed both MPP(+)-induced neuronal atrophy and cell loss. These effects were potent and of a magnitude equivalent to those achieved by a combination of brain-derived neurotrophic factor (BDNF) and glial-derived neurotrophic factor (GDNF). Oral administration of PYM50028 (10 mg/kg/day for 60 days) to MPTP-lesioned mice, commencing after a striatal impairment was evident, resulted in a significant elevation of striatal GDNF (297%) and BDNF (511%), and attenuated the loss of striatal dopaminergic transporter levels and dopaminergic neurons in the substantia nigra. PYM50028 did not inhibit monoamine oxidase B in vitro, nor did it alter brain levels of MPP(+) in vivo. PYM50028 has neuroprotective and neurorestorative potential and is in clinical development for the treatment of neurodegenerative disorders, including Parkinson's disease.


Asunto(s)
Mesencéfalo/patología , Factores de Crecimiento Nervioso/uso terapéutico , Neuronas/patología , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/prevención & control , Espirostanos/uso terapéutico , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Administración Oral , Animales , Modelos Animales de Enfermedad , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Masculino , Mesencéfalo/efectos de los fármacos , Ratones , Factores de Crecimiento Nervioso/administración & dosificación , Neuronas/efectos de los fármacos , Neurotoxinas/toxicidad , Ratas , Tirosina 3-Monooxigenasa/efectos de los fármacos , Tirosina 3-Monooxigenasa/metabolismo
4.
APL Bioeng ; 2(2): 026105, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31069302

RESUMEN

Parkinson's disease neurodegenerative brain tissue exhibits two biophysically distinct α-synuclein fiber isoforms-single stranded fibers that appear to be steric-zippers and double-stranded fibers with an undetermined structure. Herein, we describe a ß-helical homology model of α-synuclein that exhibits stability in probabilistic and Monte Carlo simulations as a candidate for stable prional dimer conformers in equilibrium with double-stranded fibers and cytotoxic pore assemblies. Molecular models of ß-helical pore assemblies are consistent with α-synucleinA53T transfected rat immunofluorescence epitope maps. Atomic force microscopy reveals that α-synuclein peptides aggregate into anisotropic fibrils lacking the density or circumference of a steric-zipper. Moreover, fibrillation was blocked by mutations designed to hinder ß-helical but not steric-zipper conformations. ß-helical species provide a structural basis for previously described biophysical properties that are incompatible with a steric-zipper, provide pathogenic mechanisms for familial human α-synuclein mutations, and offer a direct cytotoxic target for therapeutic development.

5.
Neuropharmacology ; 131: 116-127, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29197517

RESUMEN

L-DOPA-induced dyskinesia (LID) remains a significant problem in the management of Parkinson's disease (PD). In rodent and macaque models of PD, delta opioid receptor agonists have anti-parkinsonian actions while mu opioid antagonists can reduce the expression of LID. DPI-289 is a novel molecule with a unique combination of opioid receptor DAMA actions: delta agonist (Ki: 0.73 nM); mu antagonist (Ki: 12 nM). We demonstrated that DPI-289 has oral bioavailability and established its pharmacokinetic profile in both rat and primate. We hypothesised that these combined DAMA actions would provide an enhancement of L-DOPA effect without an associated increase in dyskinesia. In parkinsonian 6-OHDA lesioned rats and MPTP-lesioned macaques, DPI-289 provided anti-parkinsonian actions as monotherapy and an enhancement of L-DOPA benefit. Thus, acute administration of DPI-289 (3 mg/kg, p.o.) to 6-OHDA-lesioned rats produced a significant reduction in forelimb asymmetry (by 48%) that was maintained throughout the fifteen-day repeat-treatment period. Importantly, and in contrast to L-DOPA administration (6 mg/kg, i.p.), these benefits were not compromised by the development of abnormal involuntary movements. In the macaque, as monotherapy, DPI-289 (10 and 20 mg/kg) had significant, though incomplete, anti-parkinsonian actions lasting approximately 4 h. These benefits were not associated with dyskinesia. In fact, over the 6 h period of observation, DPI-289 (20 mg/kg) decreased parkinsonism by 19% and increased activity by 67% compared to vehicle treatment. By contrast, while high-dose L-DOPA (LDh) alone alleviated parkinsonism (for 3 h) this benefit was accompanied by significant dyskinesia that was disabling in nature. LDh provided a 50% reduction in parkinsonism over 6 h and 151% increase in activity. The combination of DPI-289 (20 mg/kg) and a low-dose of L-DOPA (LDl) provided anti-parkinsonian benefits greater than LDl alone without eliciting any significant dyskinesia. Treatment with LDl alone provided only transient statistically significant anti-parkinsonian benefit. However, the combination of LDl and DPI-289 reduced parkinsonism for 6 h (duration of monitoring), with parkinsonism being reduced by 35% and activity increased by 90% but with no increase in dyskinesia over that observed with LDl alone. Thus, DPI-289 has potential to improve the benefits of dopaminergic therapy in Parkinson's disease.


Asunto(s)
Benzamidas/uso terapéutico , Discinesia Inducida por Medicamentos/tratamiento farmacológico , Levodopa/efectos adversos , Antagonistas de Narcóticos/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Piperazinas/uso terapéutico , Receptores Opioides delta/agonistas , Receptores Opioides mu/antagonistas & inhibidores , Adrenérgicos/toxicidad , Analgésicos Opioides/farmacocinética , Analgésicos Opioides/uso terapéutico , Animales , Benzamidas/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Discinesia Inducida por Medicamentos/etiología , Femenino , Cobayas , Macaca , Masculino , Ratones , Ratones Endogámicos C57BL , Movimiento/efectos de los fármacos , Antagonistas de Narcóticos/farmacocinética , Oxidopamina/toxicidad , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/fisiopatología , Trastornos Parkinsonianos/sangre , Trastornos Parkinsonianos/tratamiento farmacológico , Piperazinas/farmacología , Ratas Sprague-Dawley , Conducto Deferente/efectos de los fármacos , Conducto Deferente/metabolismo
6.
Eur J Pharmacol ; 764: 379-384, 2015 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-26164791

RESUMEN

Mitochondrial complex I, which is the first member of the electron transport chain responsible for producing ATP, can produce reactive oxygen species and oxidative stress when it becomes dysfunctional. Complex I dysfunction and oxidative stress are strongly implicated in bipolar disorder (BD), a debilitating psychiatric disease, as is decreased levels of brain derived neurotrophic factor (BDNF) found in patients with BD, which is related to complex I activity. JNX1001, a clinical trial ready brain penetrant sapogenin, increases BDNF levels in animal models. Hence, we aimed to examine if JNX1001 can prevent complex I dysfunction-induced alterations produced by rotenone treatment in human neuroblastoma cells (SH-SY5Y). Complex I dysfunction decreased cell viability and increased protein carbonylation and nitration, confirming previous findings. Complex I dysfunction also decreased intracellular and extracellular BDNF levels. JNX1001 pre-treatment prevented complex I dysfunction-induced protein carbonylation and nitration and improved cell viability at concentrations of 30 nM and 300 nM, but more robustly at 300 nM. JNX1001 was also able to prevent decreased intracellular and extracellular BDNF levels, where it produced a ten-fold increase in intracellular BDNF levels at a concentration of 300 nM. While further studies are required to examine the neuroprotective ability of JNX1001 against alterations produced by complex I defect in more complex systems, such as in animal models, the findings of this study demonstrate the potential of JNX1001 to be used as a therapeutic agent to protect against complex I dysfunction-induced alterations that may be highly relevant to BD.


Asunto(s)
Trastorno Bipolar/tratamiento farmacológico , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Espirostanos/farmacología , Trastorno Bipolar/enzimología , Trastorno Bipolar/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Neuronas/enzimología , Neuronas/patología , Carbonilación Proteica/efectos de los fármacos , Rotenona/toxicidad , Transducción de Señal/efectos de los fármacos , Tirosina/análogos & derivados , Tirosina/metabolismo
7.
Br J Pharmacol ; 139(1): 147-55, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12746233

RESUMEN

1. The group II metabotropic glutamate (mGlu) receptor antagonist (2S,1'S,2'S)-2-(2-carboxycyclopropyl)-2-(9H-xanthen-9-yl)glycine (LY341495) also has activity at group I and III mGlu receptors at higher concentrations and can be used to discriminate between mGlu receptor subtypes. We report the antagonist action of LY341495 on glutamate receptors expressed in the neonatal rat spinal cord preparation and the use of this antagonist to investigate the group III mGlu receptor subtypes responsible for mediating the depression of synaptic transmission in the spinal cord mediated by the group III mGlu receptor agonists (S)-2-amino-4-phosphonobutanoic acid ((S)-AP4) and (1S,3R,4S)-1-aminocyclopentane-1,2,4-tricarboxylic acid (ACPT-I). 2. LY341495 antagonised mGlu receptor agonist-induced responses in the spinal cord with a rank order of potency of group II > group III > group I, which is the same as that observed in human cloned mGlu receptor cell lines. Antagonism of group II and III mGlu receptor-mediated effects were time dependent when low-nanomolar concentrations of LY341495 were used. Although the rank order of potency of LY341495 was the same on native rat and cloned human mGlu receptors, there was a compression in the selectivity between group II and III mGlu receptors, expressed in the spinal cord. 3. In agreement with a previous study on cloned ionotropic glutamate receptors 100 microM LY341495 had little or no effect on N-methyl-D-aspartate, (S)-2-amino-3-(3-hydroxy-5-methylisoxazol-4-yl) propionic acid or kainate receptor-mediated responses on motoneurones. 4. LY341495 exhibited low-nanomolar potency antagonist activity against (S)-AP4 and ACPT-I suggesting that these agonists are activating predominantly mGlu8 and that mGlu4 receptors do not play a role in modulating synaptic transmission in the pathways stimulated in the experiments described here.


Asunto(s)
Aminoácidos/farmacología , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Receptores de Glutamato Metabotrópico/fisiología , Médula Espinal/efectos de los fármacos , Xantenos/farmacología , Aminobutiratos/farmacología , Animales , Animales Recién Nacidos , Ciclopentanos/farmacología , Técnicas In Vitro , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/fisiología , Ratas , Receptores AMPA/efectos de los fármacos , Receptores de Ácido Kaínico/efectos de los fármacos , Receptores de Glutamato Metabotrópico/agonistas , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Médula Espinal/fisiología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Factores de Tiempo , Ácidos Tricarboxílicos/farmacología
8.
Br J Pharmacol ; 139(8): 1523-31, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12922940

RESUMEN

1. Three novel phenylglycine analogues; (RS)-alpha-methyl-3-chloro-4-phosphonophenylglycine (UBP1110), (RS)-alpha-methyl-3-methoxy-4-phosphonophenylglycine (UBP1111) and (RS)-alpha-methyl-3-methyl-4-phosphonophenylglycine (UBP1112) antagonised the depression of the fast component of the dorsal root-evoked ventral root potential induced by (S)-AP4 with apparent K(D) values of: 7.4+/-2.3, 5.4+/-0.6 and 5.1+/-0.3 micro M (all n=3), respectively. 2. A Schild analysis of the antagonism of (S)-AP4 induced depression of synaptic transmission by UBP1112 revealed a pA(2) value of 5.3 and a slope of 0.81+/-0.26 (n=9). 3. None of the phenylglycines tested were potent antagonists of responses mediated by group II mGlu receptors (apparent K(D) values >480 micro M). UBP1112 when tested at a concentration of 1 mM had little or no activity on (S)-3,5-DHPG-, NMDA-, AMPA- or kainate-induced responses on motoneurones. 4. UBP1110, UBP1111 and UBP1112 are at least 100-fold selective for group III over group I and II mGlu receptors expressed in the spinal cord making them the most potent, selective, antagonists yet tested at (S)-AP4 sensitive receptors in the spinal cord.


Asunto(s)
Antagonistas de Aminoácidos Excitadores/farmacología , Glicina/análogos & derivados , Glicina/farmacología , Receptores de Glutamato Metabotrópico/biosíntesis , Raíces Nerviosas Espinales/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Femenino , Glicina/química , Técnicas In Vitro , Masculino , Estructura Molecular , Ratas , Ratas Wistar , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Raíces Nerviosas Espinales/efectos de los fármacos , Relación Estructura-Actividad
9.
Eur J Pharmacol ; 649(1-3): 29-37, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-20826132

RESUMEN

To investigate the susceptibility of the group II metabotropic glutamate receptor mGlu2 to agonist-induced desensitization, the receptor was stably expressed in Chinese hamster ovary (CHO-mGlu2) or C6 glioma cells (C6-mGlu2). Exposure of CHO-mGlu2 cells to the group II mGlu receptor agonist (2S,1'S,2'S)-2-(carboxycyclopropyl)glycine (LCCG-1; 10 µM) for up to 15 h did not affect the subsequent ability of LCCG-1 to inhibit forskolin-stimulated cAMP accumulation. Similarly, in C6-mGlu2 cells, prolonged exposure to LCCG-1 also did not affect the subsequent ability of LCCG-1 to inhibit cAMP formation. In contrast, exposure of CHO-mGlu2 cells to the protein kinase C activator phorbol myristate acetate (PMA) suppressed the ability of LCCG-1 to inhibit cAMP formation. Using an in vitro model of group II mGlu receptor activity, the hemisected neonatal rat spinal cord preparation, the ability of the selective group II agonist (2R,4R)-4-aminopyrrolidine-2,4-dicarboxylate ((2R,4R)-APDC) to depress the fast component of the dorsal root-evoked ventral root potential (fDR-VRP) did not desensitize when applied for up to 2 h. Together these results indicate that in contrast to most G protein-coupled receptors, the mGlu2 receptor is resistant to agonist-induced homologous desensitization, and that in vitro data suggests that resistance to desensitization is a physiologically relevant property of this mGlu receptor subtype.


Asunto(s)
Agonistas de Aminoácidos Excitadores/farmacología , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/metabolismo , Proteína Quinasa C/metabolismo , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/metabolismo , Adenilil Ciclasas/metabolismo , Aminoácidos Dicarboxílicos/antagonistas & inhibidores , Aminoácidos Dicarboxílicos/farmacología , Animales , Animales Recién Nacidos , Células CHO , Línea Celular , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Activadores de Enzimas/farmacología , Potenciales Evocados/efectos de los fármacos , Técnicas In Vitro , Proteínas del Tejido Nervioso/genética , Neuronas/efectos de los fármacos , Neuronas/fisiología , Ratas , Ratas Wistar , Receptor del Glutamato Metabotropico 5 , Receptores de Glutamato Metabotrópico/genética , Raíces Nerviosas Espinales/efectos de los fármacos , Raíces Nerviosas Espinales/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA