Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Neurobiol Dis ; 35(3): 399-405, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19501651

RESUMEN

High frequency stimulation (HFS) of the internal pallidum is effective for the treatment of dystonia. Only few studies have investigated the effects of stimulation on the activity of the cortex-basal ganglia network. We here assess within this network the effect of entopeduncular nucleus (EP) HFS on the expression of c-Fos and cytochrome oxidase subunit I (COI) in the dt(sz)-hamster, a well-characterized model of paroxysmal dystonia. In dt(sz)-hamsters, we identified abnormal activity in motor cortex, basal ganglia and thalamus. These structures have already been linked to the pathophysiology of human dystonia. EP-HFS (i) increased striatal c-Fos expression in controls and dystonic hamsters and (ii) reduced thalamic c-Fos expression in dt(sz)-hamsters. EP-HFS had no effect on COI expression. The present results suggest that EP-HFS induces a new network activity state which may improve information processing and finally reduces the severity of dystonic attacks in dt(sz)-hamsters.


Asunto(s)
Ganglios Basales/fisiopatología , Corteza Cerebral/fisiopatología , Estimulación Encefálica Profunda , Distonía/terapia , Núcleo Entopeduncular/fisiopatología , Animales , Encéfalo/fisiopatología , Cuerpo Estriado/fisiopatología , Cricetinae , Distonía/fisiopatología , Estimulación Eléctrica , Complejo IV de Transporte de Electrones/metabolismo , Femenino , Inmunohistoquímica , Masculino , Vías Nerviosas/fisiopatología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Índice de Severidad de la Enfermedad
2.
Ultrastruct Pathol ; 32(5): 178-83, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18958789

RESUMEN

Human telomerase comprises a catalytic protein subunit (hTERT) and an RNA subunit (hTR). Telomerase extends chromosome ends in compensation for the attrition of the telomeres during replication. In this work, the authors explore the expression of hTERT and hTR in neutrophils, respectively by immunochemistry techniques and in situ hybridization. hTERT was strongly expressed in neutrophils cytoplasm. The ultrastructural study showed that the gold particles were not associated with specific organelles but scattered in the cytosol. hTR was not expressed. hTERT is expressed in the cytoplasm of neutrophils, but its roles-eventually extratelomeric effects-remain to be elucidated.


Asunto(s)
Citoplasma/enzimología , Microscopía Inmunoelectrónica , Neutrófilos/enzimología , Telomerasa/análisis , Citoplasma/ultraestructura , Humanos , Hibridación in Situ , Hígado/enzimología , Hígado/ultraestructura , Neutrófilos/ultraestructura , ARN , ARN Largo no Codificante , ARN no Traducido/análisis , Vejiga Urinaria/enzimología , Vejiga Urinaria/ultraestructura
3.
Biol Psychiatry ; 61(7): 836-44, 2007 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-16950226

RESUMEN

BACKGROUND: A role for enhanced opioid peptide transmission has been suggested in the genesis of levodopa-induced dyskinesia. However, basal ganglia nuclei other than the striatum have not been regarded as potential sources, and the opioid precursors have never been quantified simultaneously with the levels of opioid receptors at the peak of dyskinesia severity. METHODS: The levels of messenger RNA (mRNA) encoding the opioid precursors preproenkephalin-A and preproenkephalin-B in the striatum and the subthalamic nucleus and the levels of mu, delta, and kappa opioid receptors were measured within the basal ganglia of four groups of nonhuman primates killed at the peak of effect: normal, parkinsonian, parkinsonian chronically-treated with levodopa without exhibiting dyskinesia, and parkinsonian chronically-treated with levodopa showing overt dyskinesia. RESULTS: Dyskinesia are associated with reduction in opioid receptor binding and specifically of kappa and mu receptor binding in the globus pallidus internalis (GPi), the main output structure of the basal ganglia. This decrease was correlated with enhancement of the expression of preproenkephalin-B mRNA but not that of preproenkephalin-A in the striatum and the subthalamic nucleus. CONCLUSIONS: Abnormal transmission of preproenkephalin-B-derived opioid coming from the striatum and the subthalamic nucleus converges upon GPi at the peak of dose to induce levodopa-induced dyskinesia.


Asunto(s)
Cuerpo Estriado/metabolismo , Discinesia Inducida por Medicamentos , Encefalinas/metabolismo , Regulación de la Expresión Génica/fisiología , Precursores de Proteínas/metabolismo , Receptores Opioides/metabolismo , Núcleo Subtalámico/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Análisis de Varianza , Animales , Antiparkinsonianos/administración & dosificación , Antiparkinsonianos/efectos adversos , Interacciones Farmacológicas , Discinesia Inducida por Medicamentos/metabolismo , Discinesia Inducida por Medicamentos/patología , Discinesia Inducida por Medicamentos/fisiopatología , Encefalinas/genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hibridación in Situ/métodos , Levodopa/administración & dosificación , Levodopa/efectos adversos , Macaca fascicularis , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/tratamiento farmacológico , Precursores de Proteínas/genética , ARN Mensajero/metabolismo , Ensayo de Unión Radioligante/métodos , Análisis de Regresión
4.
J Neurosci ; 25(8): 2102-7, 2005 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-15728850

RESUMEN

Dyskinesia represents a debilitating complication of L-3,4-dihydroxyphenylalanine (L-dopa) therapy for Parkinson's disease. Such motor manifestations are attributed to pathological activity in the motor parts of basal ganglia. However, because consistent funneling of information takes place between the sensorimotor, limbic, and associative basal ganglia domains, we hypothesized that nonmotor domains play a role in these manifestations. Here we report the changes in 2-deoxyglucose (2-DG) accumulation in the sensorimotor, limbic, and associative domains of basal ganglia and thalamic nuclei of four groups of nonhuman primates: normal, parkinsonian, parkinsonian chronically treated with L-dopa without exhibiting dyskinesia, and parkinsonian chronically treated with L-dopa and exhibiting overt dyskinesia. Although nondyskinetic animals display a rather normalized metabolic activity, dyskinetic animals are distinguished by significant changes in 2-DG accumulation in limbic- and associative-related structures and not simply in sensorimotor-related ones, suggesting that dyskinesia is linked to a pathological processing of limbic and cognitive information. We propose that these metabolic changes reflect the underlying neural mechanisms of not simply motor dyskinesias but also affective, motivational, and cognitive disorders associated with long-term exposure to L-dopa.


Asunto(s)
Ganglios Basales/fisiopatología , Discinesia Inducida por Medicamentos/fisiopatología , Levodopa/toxicidad , Sistema Límbico/fisiopatología , Animales , Ganglios Basales/química , Desoxiglucosa/farmacocinética , Discinesia Inducida por Medicamentos/metabolismo , Femenino , Globo Pálido/química , Globo Pálido/fisiopatología , Levodopa/uso terapéutico , Sistema Límbico/química , Macaca fascicularis , Corteza Motora/química , Corteza Motora/fisiopatología , Trastornos Parkinsonianos/tratamiento farmacológico , Trastornos Parkinsonianos/fisiopatología , Núcleos Septales/química , Núcleos Septales/fisiopatología , Corteza Somatosensorial/química , Corteza Somatosensorial/fisiopatología , Sustancia Negra/química , Sustancia Negra/fisiopatología , Núcleo Subtalámico/química , Núcleo Subtalámico/fisiopatología
5.
PLoS One ; 5(10): e13306, 2010 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-20967255

RESUMEN

BACKGROUND: The A11 diencephalospinal pathway is crucial for sensorimotor integration and pain control at the spinal cord level. When disrupted, it is thought to be involved in numerous painful conditions such as restless legs syndrome and migraine. Its anatomical organization, however, remains largely unknown in the non-human primate (NHP). We therefore characterized the anatomy of this pathway in the NHP. METHODS AND FINDINGS: In situ hybridization of spinal dopamine receptors showed that D1 receptor mRNA is absent while D2 and D5 receptor mRNAs are mainly expressed in the dorsal horn and D3 receptor mRNA in both the dorsal and ventral horns. Unilateral injections of the retrograde tracer Fluoro-Gold (FG) into the cervical spinal enlargement labeled A11 hypothalamic neurons quasi-exclusively among dopamine areas. Detailed immunohistochemical analysis suggested that these FG-labeled A11 neurons are tyrosine hydroxylase-positive but dopa-decarboxylase and dopamine transporter-negative, suggestive of a L-DOPAergic nucleus. Stereological cell count of A11 neurons revealed that this group is composed by 4002±501 neurons per side. A 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) intoxication with subsequent development of a parkinsonian syndrome produced a 50% neuronal cell loss in the A11 group. CONCLUSION: The diencephalic A11 area could be the major source of L-DOPA in the NHP spinal cord, where it may play a role in the modulation of sensorimotor integration through D2 and D3 receptors either directly or indirectly via dopamine formation in spinal dopa-decarboxylase-positives cells.


Asunto(s)
Diencéfalo/anatomía & histología , Primates/anatomía & histología , Médula Espinal/anatomía & histología , Animales , Diencéfalo/metabolismo , Dopamina/metabolismo , Inmunohistoquímica , Hibridación in Situ , ARN Mensajero/genética , Receptores Dopaminérgicos/genética , Médula Espinal/metabolismo
6.
Sci Transl Med ; 2(28): 28ra28, 2010 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-20410529

RESUMEN

Parkinson's disease is caused primarily by degeneration of brain dopaminergic neurons in the substantia nigra and the consequent deficit of dopamine in the striatum. Dopamine replacement therapy with the dopamine precursor l-dopa is the mainstay of current treatment. After several years, however, the patients develop l-dopa-induced dyskinesia, or abnormal involuntary movements, thought to be due to excessive signaling via dopamine receptors. G protein-coupled receptor kinases (GRKs) control desensitization of dopamine receptors. We found that dyskinesia is attenuated by lentivirus-mediated overexpression of GRK6 in the striatum in rodent and primate models of Parkinson's disease. Conversely, reduction of GRK6 concentration by microRNA delivered with lentiviral vector exacerbated dyskinesia in parkinsonian rats. GRK6 suppressed dyskinesia in monkeys without compromising the antiparkinsonian effects of l-dopa and even prolonged the antiparkinsonian effect of a lower dose of l-dopa. Our finding that increased availability of GRK6 ameliorates dyskinesia and increases duration of the antiparkinsonian action of l-dopa suggests a promising approach for controlling both dyskinesia and motor fluctuations in Parkinson's disease.


Asunto(s)
Discinesias/complicaciones , Discinesias/prevención & control , Quinasas de Receptores Acoplados a Proteína-G/uso terapéutico , Terapia Genética , Lentivirus/genética , Trastornos Parkinsonianos/complicaciones , Trastornos Parkinsonianos/terapia , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Animales , Antiparkinsonianos/farmacología , Antiparkinsonianos/uso terapéutico , Conducta Animal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Endocitosis/efectos de los fármacos , Quinasas de Receptores Acoplados a Proteína-G/genética , Técnicas de Silenciamiento del Gen , Humanos , Levodopa , Macaca , Oxidopamina/farmacología , Trastornos Parkinsonianos/tratamiento farmacológico , Trastornos Parkinsonianos/genética , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D2/metabolismo , Rotación , Transducción de Señal/efectos de los fármacos
7.
Biol Psychiatry ; 66(6): 554-61, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19481198

RESUMEN

BACKGROUND: Involuntary movements, or dyskinesia, represent a debilitating complication of dopamine replacement therapy for Parkinson disease (PD). The transcription factor DeltaFosB accumulates in the denervated striatum and dimerizes primarily with JunD upon repeated L-3,4-dihydroxyphenylalanine (L-DOPA) administration. Previous studies in rodents have shown that striatal DeltaFosB levels accurately predict dyskinesia severity and indicate that this transcription factor may play a causal role in the dyskinesia sensitization process. METHODS: We asked whether the correlation previously established in rodents extends to the best nonhuman primate model of PD, the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned macaque. We used western blotting and quantitative polymerase chain reaction (PCR) to compare DeltaFosB protein and messenger RNA (mRNA) levels across two subpopulations of macaques with differential dyskinesia severity. Second, we tested the causal implication of DeltaFosB in this primate model. Serotype 2 adeno-associated virus (AAV2) vectors were used to overexpress, within the motor striatum, either DeltaFosB or DeltaJunD, a truncated variant of JunD lacking a transactivation domain and therefore acting as a dominant negative inhibitor of DeltaFosB. RESULTS: A linear relationship was observed between endogenous striatal levels of DeltaFosB and the severity of dyskinesia in Parkinsonian macaques treated with L-DOPA. Viral overexpression of DeltaFosB did not alter dyskinesia severity in animals previously rendered dyskinetic, whereas the overexpression of DeltaJunD dramatically dropped the severity of this side effect of L-DOPA without altering the antiparkinsonian activity of the treatment. CONCLUSIONS: These results establish a mechanism of dyskinesia induction and maintenance by L-DOPA and validate a strategy, with strong translational potential, to deprime the L-DOPA-treated brain.


Asunto(s)
Antiparkinsonianos/efectos adversos , Benserazida/efectos adversos , Cuerpo Estriado/metabolismo , Discinesia Inducida por Medicamentos/patología , Discinesia Inducida por Medicamentos/terapia , Regulación de la Expresión Génica/fisiología , Levodopa/efectos adversos , Proteínas Proto-Oncogénicas c-jun/metabolismo , Análisis de Varianza , Animales , Cuerpo Estriado/diagnóstico por imagen , Modelos Animales de Enfermedad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Combinación de Medicamentos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Terapia Genética/métodos , Vectores Genéticos/fisiología , Proteínas Fluorescentes Verdes/genética , Humanos , Radioisótopos de Yodo , Modelos Lineales , Intoxicación por MPTP/diagnóstico por imagen , Intoxicación por MPTP/tratamiento farmacológico , Intoxicación por MPTP/patología , Macaca fascicularis , Masculino , Nortropanos , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-jun/genética , ARN Mensajero/metabolismo , Cintigrafía
8.
Biol Psychiatry ; 65(6): 518-26, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18947822

RESUMEN

BACKGROUND: Chronic L-3,4-dihydroxyphenylalanine (L-DOPA) treatment of Parkinson's disease (PD) leads to debilitating involuntary movements, termed L-DOPA-induced dyskinesia. Striatofugal medium spiny neurons (MSN) lose their dendritic spines and cortico-striatal glutamatergic synapses in PD and in experimental models of DA depletion. This loss of connectivity is triggered by a dysregulation of intraspine Cav1.3 L-type Ca2+ channels. Here we address the possible implication of DA denervation-induced spine pruning in the development of L-DOPA-induced dyskinesia. METHODS: The L-type Ca2+ antagonist, isradipine was subcutaneously delivered to rats at the doses of .05, .1, or .2 mg/kg/day, for 4 weeks, starting the day after a unilateral nigrostriatal 6-hydroxydopamine (6-OHDA) lesion. Fourteen days later, L-DOPA treatment was initiated. RESULTS: Isradipine-treated animals displayed a dose-dependent reduction in L-DOPA-induced rotational behavior and abnormal involuntary movements. Dendritic spine counting at electron microscopy level showed that isradipine (.2 mg/kg/day) prevented the 6-OHDA-induced spine loss and normalized preproenkephalin-A messenger RNA expression. Involuntary movements were not reduced when isradipine treatment was started concomitantly with L-DOPA. CONCLUSIONS: These results indicate that isradipine, at a therapeutically relevant dose, might represent a treatment option for preventing L-DOPA-induced dyskinesia in PD.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/efectos de los fármacos , Discinesia Inducida por Medicamentos/prevención & control , Isradipino/uso terapéutico , Levodopa/efectos adversos , Simpaticolíticos/administración & dosificación , Animales , Bloqueadores de los Canales de Calcio/administración & dosificación , Cerebro/metabolismo , Cerebro/ultraestructura , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/ultraestructura , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Discinesia Inducida por Medicamentos/metabolismo , Encefalinas/metabolismo , Isradipino/administración & dosificación , Isradipino/farmacología , Levodopa/farmacología , Masculino , Actividad Motora/efectos de los fármacos , Nimodipina/farmacología , Oxidopamina , Precursores de Proteínas/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar
9.
Eur J Neurosci ; 22(1): 283-7, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16029219

RESUMEN

The extent of nigrostriatal denervation is presumed to play a role in the genesis of levodopa-induced dyskinesia. Yet some parkinsonian patients who have been treated over a long period do not develop dyskinesia, raising the possibility that the pattern of denervation is as important as the extent of lesioning as a risk factor. Here we study the extent and pattern of nigrostriatal denervation in a homogeneous population of parkinsonian macaque monkeys chronically treated with levodopa. Based on the characteristics of the lesioning, non-dyskinetic animals could not be differentiated from those with dyskinesia. Indeed, the number of tyrosine-hydroxylase (TH)-immunopositive neurons in the substantia nigra pars compacta, striatal dopamine transporter (DAT) binding and TH immunostaining, as well as the overall TH striatal content measured by Western blotting were identical. Moreover, the patterns of lesioning assessed by a detailed analysis of the TH- and DAT-immunopositive striatal fibers were comparable in all functional quadrants and at all rostro-caudal levels considered. These data indicate that neither the extent nor the pattern of nigrostriatal lesioning are sufficient to explain the occurrence of levodopa-induced dyskinesia.


Asunto(s)
Discinesia Inducida por Medicamentos/patología , Levodopa/efectos adversos , Vías Nerviosas/patología , Trastornos Parkinsonianos/patología , Sustancia Negra/patología , Animales , Antiparkinsonianos/efectos adversos , Recuento de Células , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Cuerpo Estriado/fisiopatología , Modelos Animales de Enfermedad , Dopamina/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática , Discinesia Inducida por Medicamentos/fisiopatología , Femenino , Inmunohistoquímica , Macaca fascicularis , Glicoproteínas de Membrana/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/patología , Degeneración Nerviosa/fisiopatología , Proteínas del Tejido Nervioso/metabolismo , Vías Nerviosas/metabolismo , Vías Nerviosas/fisiopatología , Neuronas/metabolismo , Neuronas/patología , Trastornos Parkinsonianos/tratamiento farmacológico , Trastornos Parkinsonianos/fisiopatología , Sustancia Negra/metabolismo , Sustancia Negra/fisiopatología , Tirosina 3-Monooxigenasa/metabolismo
10.
Ann Neurol ; 57(1): 17-26, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15514976

RESUMEN

Involuntary movements, or dyskinesia, represent a debilitating complication of levodopa therapy for Parkinson's disease. Although changes affecting D(1) and D(2) dopamine receptors have been studied in association with this condition, no causal relationship has yet been established. Taking advantage of a monkey brain bank constituted to study levodopa-induced dyskinesia, we report changes affecting D(1) and D(2) dopamine receptors within the striatum of normal, parkinsonian, nondyskinetic levodopa-treated parkinsonian, and dyskinetic levodopa-treated parkinsonian animals. Whereas D(1) receptor expression itself is not related to dyskinesia, D(1) sensitivity per D(1) receptor measured by D(1) agonist-induced [(35)S]GTPgammaS binding is linearly related to dyskinesia. Moreover, the striata of dyskinetic animals show higher levels of cyclin-dependent kinase 5 (Cdk5) and of the dopamine- and cAMP-regulated phosphoprotein of 32kDa (DARPP-32). Our data suggest that levodopa-induced dyskinesia results from increased dopamine D(1) receptor-mediated transmission at the level of the direct pathway.


Asunto(s)
Antiparkinsonianos/efectos adversos , Discinesia Inducida por Medicamentos/etiología , Levodopa/efectos adversos , Receptores de Dopamina D1/metabolismo , Transducción de Señal/efectos de los fármacos , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Análisis de Varianza , Animales , Autorradiografía/métodos , Conducta Animal , Western Blotting/métodos , Quinasa 5 Dependiente de la Ciclina , Quinasas Ciclina-Dependientes , Modelos Animales de Enfermedad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática , Fosfoproteína 32 Regulada por Dopamina y AMPc , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Discinesia Inducida por Medicamentos/metabolismo , Femenino , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Inmunohistoquímica/métodos , Hibridación in Situ/métodos , Isótopos/farmacocinética , Macaca fascicularis , Glicoproteínas de Membrana/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Actividad Motora/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Nortropanos/farmacocinética , Trastornos Parkinsonianos/tratamiento farmacológico , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/fisiopatología , Fosfoproteínas/metabolismo , Ensayo de Unión Radioligante/métodos , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/genética , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Factores de Tiempo , Tirosina 3-Monooxigenasa/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda