Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Purinergic Signal ; 16(3): 379-387, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32725400

RESUMEN

Parkinson's disease (PD) signs and symptoms regularly include tremor. Interestingly, the nucleoside guanosine (GUO) has already proven to be effective in reducing reserpine-induced tremulous jaw movements (TJMs) in rodent models, thus becoming a promising antiparkinsonian drug. Here, we aimed at revealing the mechanism behind GUO antiparkinsonian efficacy by assessing the role of adenosine A1 and A2A receptors (A1R and A2AR) on GUO-mediated anti-tremor effects in the reserpinized mouse model of PD. Reserpinized mice showed elevated reactive oxygen species (ROS) production and cellular membrane damage in striatal slices assessed ex vivo and GUO treatment reversed ROS production. Interestingly, while the simultaneous administration of sub-effective doses of GUO (5 mg/kg) and SCH58261 (0.01 mg/kg), an A2AR antagonist, precluded reserpine-induced TJMs, these were ineffective on reverting ROS production in ex vivo experiments. Importantly, GUO was able to reduce TJM and ROS production in reserpinized mouse lacking the A2AR, thus suggesting an A2AR-independent mechanism of GUO-mediated effects. Conversely, the administration of DPCPX (0.75 mg/kg), an A1R antagonist, completely abolished both GUO-mediated anti-tremor effects and blockade of ROS production. Overall, these results indicated that GUO anti-tremor and antioxidant effects in reserpinized mice were A1R dependent but A2AR independent, thus suggesting a differential participation of adenosine receptors in GUO-mediated effects.


Asunto(s)
Guanosina/uso terapéutico , Enfermedad de Parkinson Secundaria/metabolismo , Receptor de Adenosina A1/metabolismo , Receptor de Adenosina A2A/metabolismo , Temblor/metabolismo , Antagonistas del Receptor de Adenosina A1/farmacología , Antagonistas del Receptor de Adenosina A2 , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Guanosina/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Ratones , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Temblor/inducido químicamente , Temblor/tratamiento farmacológico , Xantinas/farmacología
2.
Neuropathol Appl Neurobiol ; 43(5): 373-392, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28117912

RESUMEN

AIMS: The present study analyses molecular characteristics of the locus coeruleus (LC) and projections to the amygdala and hippocampus at asymptomatic early and middle Braak stages of neurofibrillary tangle (NFT) pathology. METHODS: Immunohistochemistry, whole-transcriptome arrays and RT-qPCR in LC and western blotting in hippocampus and amygdala in a cohort of asymptomatic individuals at stages I-IV of NFT pathology were used. RESULTS: NFTs in the LC increased in parallel with colocalized expression of tau kinases, increased neuroketal adducts and decreased superoxide dismutase 1 in neurons with hyperphosphorylated tau and decreased voltage-dependent anion channel in neurons containing truncated tau were found. These were accompanied by increased microglia and AIF1, CD68, PTGS2, IL1ß, IL6 and TNF-α gene expression. Whole-transcriptome arrays revealed upregulation of genes coding for proteins associated with heat shock protein binding and genes associated with ATP metabolism and downregulation of genes coding for DNA-binding proteins and members of the small nucleolar RNAs family, at stage IV when compared with stage I. Tyrosine hydroxylase (TH) immunoreactivity was preserved in neurons of the LC, but decreased TH and increased α2A adrenergic receptor protein levels were found in the hippocampus and the amygdala. CONCLUSIONS: Complex alteration of several metabolic pathways occurs in the LC accompanying NFT formation at early and middle asymptomatic stages of NFT pathology. Dopaminergic/noradrenergic denervation and increased expression of α2A adrenergic receptor in the hippocampus and amygdala occur at first stage of NFT pathology, suggesting compensatory activation in the face of decreased adrenergic input occurring before clinical evidence of cognitive impairment and depression.


Asunto(s)
Locus Coeruleus/metabolismo , Locus Coeruleus/patología , Ovillos Neurofibrilares/patología , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad
3.
Genes Brain Behav ; 17(4): e12432, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29053217

RESUMEN

G protein-coupled receptors (GPCR) exhibit the ability to form receptor complexes that include molecularly different GPCR (ie, GPCR heteromers), which endow them with singular functional and pharmacological characteristics. The relative expression of GPCR heteromers remains a matter of intense debate. Recent studies support that adenosine A2A receptors (A2A R) and dopamine D2 receptors (D2 R) predominantly form A2A R-D2 R heteromers in the striatum. The aim of the present study was evaluating the behavioral effects of pharmacological manipulation and genetic blockade of A2A R and D2 R within the frame of such a predominant striatal heteromeric population. First, in order to avoid possible strain-related differences, a new D2 R-deficient mouse with the same genetic background (CD-1) than the A2A R knock-out mouse was generated. Locomotor activity, pre-pulse inhibition (PPI) and drug-induced catalepsy were then evaluated in wild-type, A2A R and D2 R knock-out mice, with and without the concomitant administration of either the D2 R agonist sumanirole or the A2A R antagonist SCH442416. SCH442416-mediated locomotor effects were demonstrated to be dependent on D2 R signaling. Similarly, a significant dependence on A2A R signaling was observed for PPI and for haloperidol-induced catalepsy. The results could be explained by the existence of one main population of striatal postsynaptic A2A R-D2 R heteromers, which may constitute a relevant target for the treatment of Parkinson's disease and other neuropsychiatric disorders.


Asunto(s)
Conducta Animal/fisiología , Cuerpo Estriado/fisiología , Receptor de Adenosina A2A/fisiología , Receptores de Dopamina D2/fisiología , Adenosina/metabolismo , Antagonistas del Receptor de Adenosina A2/farmacología , Animales , Conducta Animal/efectos de los fármacos , Bencimidazoles/farmacología , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Femenino , Locomoción/efectos de los fármacos , Locomoción/fisiología , Masculino , Ratones , Ratones Noqueados , Neostriado/metabolismo , Pirazoles/farmacología , Pirimidinas/farmacología , Receptor de Adenosina A2A/metabolismo , Receptores de Dopamina D2/metabolismo , Transducción de Señal/efectos de los fármacos
4.
CNS Neurol Disord Drug Targets ; 12(8): 1128-42, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24040811

RESUMEN

Metabotropic glutamate (mGlu) receptors are G protein-coupled receptors expressed primarily on neurons and glial cells modulating the effects of glutamatergic neurotransmission. The pharmacological manipulation of these receptors has been postulated to be valuable in the management of some neurological disorders. Accordingly, the targeting of mGlu5 receptors as a therapeutic approach for Parkinson's disease (PD) has been proposed, especially to manage the adverse symptoms associated to chronic treatment with classical PD drugs. Thus, the specific pharmacological blocking of mGlu5 receptors constitutes one of the most attractive non-dopaminergic-based strategies for PD management in general and for the L-DOPA-induced dyskinesia (LID) in particular. Overall, we provide here an update of the current state of the art of these mGlu5 receptor-based approaches that are under clinical study as agents devoted to alleviate PD symptoms.


Asunto(s)
Antiparkinsonianos/administración & dosificación , Cuerpo Estriado/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Receptor del Glutamato Metabotropico 5/metabolismo , Animales , Ensayos Clínicos como Asunto/métodos , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/patología , Humanos , Enfermedad de Parkinson/patología , Receptor del Glutamato Metabotropico 5/genética
5.
CNS Neurosci Ther ; 16(3): e18-42, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20345970

RESUMEN

Adenosine-dopamine interactions in the central nervous system (CNS) have been studied for many years in view of their relevance for disorders of the CNS and their treatments. The discovery of adenosine and dopamine receptor containing receptor mosaics (RM, higher-order receptor heteromers) in the striatum opened up a new understanding of these interactions. Initial findings indicated the existence of A(2A)R-D(2)R heterodimers and A(1)R-D(1)R heterodimers in the striatum that were followed by indications for the existence of striatal A(2A)R-D(3)R and A(2A)R-D(4)R heterodimers. Of particular interest was the demonstration that antagonistic allosteric A(2A)-D(2) and A(1)-D(1) receptor-receptor interactions take place in striatal A(2A)R-D(2)R and A(1)R-D(1)R heteromers. As a consequence, additional characterization of these heterodimers led to new aspects on the pathophysiology of Parkinson's disease (PD), schizophrenia, drug addiction, and l-DOPA-induced dyskinesias relevant for their treatments. In fact, A(2A)R antagonists were introduced in the symptomatic treatment of PD in view of the discovery of the antagonistic A(2A)R-D(2)R interaction in the dorsal striatum that leads to reduced D(2)R recognition and G(i/o) coupling in striato-pallidal GABAergic neurons. In recent years, indications have been obtained that A(2A)R-D(2)R and A(1)R-D(1)R heteromers do not exist as heterodimers, rather as RM. In fact, A(2A)-CB(1)-D(2) RM and A(2A)-D(2)-mGlu(5) RM have been discovered using a sequential BRET-FRET technique and by using the BRET technique in combination with bimolecular fluorescence complementation. Thus, other pathogenic mechanisms beside the well-known alterations in the release and/or decoding of dopamine in the basal ganglia and limbic system are involved in PD, schizophrenia and drug addiction. In fact, alterations in the stoichiometry and/or topology of A(2A)-CB(1)-D(2) and A(2A)-D(2)-mGlu5 RM may play a role. Thus, the integrative receptor-receptor interactions in these RM give novel aspects on the pathophysiology and treatment strategies, based on combined treatments, for PD, schizophrenia, and drug addiction.


Asunto(s)
Adenosina/metabolismo , Enfermedades del Sistema Nervioso Central/fisiopatología , Enfermedades del Sistema Nervioso Central/terapia , Dopamina/metabolismo , Animales , Interacciones Farmacológicas , Humanos , Modelos Biológicos , Modelos Moleculares , Receptores Dopaminérgicos/fisiología , Receptores Purinérgicos P1/fisiología , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA