Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Neurol Sci ; 435: 120193, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35259650

RESUMEN

There has been a long history of lesioning procedures to treat tremor associated with both essential tremor (ET) and Parkinson's disease (PD). These include radiofrequency (RF) thalamotomy, gamma knife radiosurgical (GKRS) thalamotomy, and magnetic resonance-guided focused ultrasound (MRgFUS). In this review, we summarize the clinical studies of lesioning procedures for tremor focusing on these ablative therapies for ET and tremor-predominant PD (TDPD). We then consider clinical treatment variables that influence decision-making regarding ablative therapies versus consideration of deep brain stimulation (DBS) and conclude with ongoing and future studies. This article is part of the Special Issue "Tremor" edited by Daniel D. Truong, Mark Hallett, and Aasef Shaikh.


Asunto(s)
Temblor Esencial , Enfermedad de Parkinson , Radiocirugia , Temblor Esencial/diagnóstico por imagen , Temblor Esencial/cirugía , Humanos , Imagen por Resonancia Magnética/métodos , Enfermedad de Parkinson/cirugía , Enfermedad de Parkinson/terapia , Tálamo/diagnóstico por imagen , Tálamo/cirugía , Resultado del Tratamiento , Temblor/diagnóstico por imagen , Temblor/terapia
2.
Neurotherapeutics ; 18(2): 1175-1187, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33594533

RESUMEN

Fragile X syndrome (FXS) is a genetic neurodevelopmental syndrome characterized by increased anxiety, repetitive behaviors, social communication deficits, delayed language development, and abnormal sensory processing. Recently, we have identified electroencephalographic (EEG) biomarkers that are conserved between the mouse model of FXS (Fmr1 KO mice) and humans with FXS. In this study, we test a specific candidate mechanism for engagement of multielectrode array (MEA) EEG biomarkers in the FXS mouse model. We administered TAK-063, a potent, selective, and orally active phosphodiesterase 10A (PDE10A) inhibitor, to Fmr1 KO mice, and examined its effects on MEA EEG biomarkers. We demonstrate significant dose-related amelioration of inter-trial phase coherence (ITPC) to temporally modulated auditory stimuli by TAK-063 in Fmr1 KO mice. Our data suggest that TAK-063 improves cortical auditory stimulus processing in Fmr1 KO mice, without significantly depressing baseline EEG power or causing any noticeable sedation or behavioral side effects. Thus, the PDE10A inhibitor TAK-063 has salutary effects on normalizing EEG biomarkers in a mouse model of FXS and should be pursued in further translational treatment development.


Asunto(s)
Estimulación Acústica/efectos adversos , Electroencefalografía/efectos de los fármacos , Síndrome del Cromosoma X Frágil/tratamiento farmacológico , Inhibidores de Fosfodiesterasa/uso terapéutico , Hidrolasas Diéster Fosfóricas , Pirazoles/uso terapéutico , Piridazinas/uso terapéutico , Animales , Electroencefalografía/métodos , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Inhibidores de Fosfodiesterasa/farmacología , Pirazoles/farmacología , Piridazinas/farmacología
3.
J Neurochem ; 155(5): 538-558, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32374912

RESUMEN

Individuals with Fragile X Syndrome (FXS) and autism spectrum disorder (ASD) exhibit cognitive impairments, social deficits, increased anxiety, and sensory hyperexcitability. Previously, we showed that elevated levels of matrix metalloproteinase-9 (MMP-9) may contribute to abnormal development of parvalbumin (PV) interneurons and perineuronal nets (PNNs) in the developing auditory cortex (AC) of Fmr1 knock-out (KO) mice, which likely underlie auditory hypersensitivity. Thus, MMP-9 may serve as a potential target for treatment of auditory hypersensitivity in FXS. Here, we used the MMP-2/9 inhibitor, SB-3CT, to pharmacologically inhibit MMP-9 activity during a specific developmental period and to test whether inhibition of MMP-9 activity reverses neural oscillation deficits and behavioral impairments by enhancing PNN formation around PV cells in Fmr1 KO mice. Electroencephalography (EEG) was used to measure resting state and sound-evoked electrocortical activity in auditory and frontal cortices of postnatal day (P)22-23 male mice before and one-day after treatment with SB-3CT (25 mg/kg) or vehicle. At P27-28, animal behaviors were tested to measure the effects of the treatment on anxiety and hyperactivity. Results show that acute inhibition of MMP-9 activity improved evoked synchronization to auditory stimuli and ameliorated mouse behavioral deficits. MMP-9 inhibition enhanced PNN formation, increased PV levels and TrkB phosphorylation yet reduced Akt phosphorylation in the AC of Fmr1 KO mice. Our results show that MMP-9 inhibition during early postnatal development is beneficial in reducing some auditory processing deficits in the FXS mouse model and may serve as a candidate therapeutic for reversing sensory hypersensitivity in FXS and possibly other ASDs.


Asunto(s)
Estimulación Acústica/métodos , Percepción Auditiva/fisiología , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Compuestos Heterocíclicos con 1 Anillo/farmacología , Metaloproteinasa 9 de la Matriz/metabolismo , Red Nerviosa/metabolismo , Sulfonas/farmacología , Animales , Animales Recién Nacidos , Corteza Auditiva/efectos de los fármacos , Corteza Auditiva/metabolismo , Percepción Auditiva/efectos de los fármacos , Electroencefalografía/efectos de los fármacos , Electroencefalografía/métodos , Inhibidores Enzimáticos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Red Nerviosa/efectos de los fármacos , Nervios Periféricos/crecimiento & desarrollo , Nervios Periféricos/metabolismo
4.
Neurobiol Dis ; 138: 104794, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32036032

RESUMEN

Fragile X Syndrome (FXS) is a leading known genetic cause of intellectual disability with symptoms that include increased anxiety and social and sensory processing deficits. Recent EEG studies in humans with FXS have identified neural oscillation deficits that include increased resting state gamma power, increased amplitude of auditory evoked potentials, and reduced inter-trial phase coherence of sound-evoked gamma oscillations. Identification of comparable EEG biomarkers in mouse models of FXS could facilitate the pre-clinical to clinical therapeutic pipeline. However, while human EEG studies have involved 128-channel scalp EEG acquisition, no mouse studies have been performed with more than three EEG channels. In the current study, we employed a recently developed 30-channel mouse multielectrode array (MEA) system to record and analyze resting and stimulus-evoked EEG signals in WT vs. Fmr1 KO mice. Using this system, we now report robust MEA-derived phenotypes including higher resting EEG power, altered event-related potentials (ERPs) and reduced inter-trial phase coherence to auditory chirp stimuli in Fmr1 KO mice that are remarkably similar to those reported in humans with FXS. We propose that the MEA system can be used for: (i) derivation of higher-level EEG parameters; (ii) EEG biomarkers for drug testing; and (ii) mechanistic studies of FXS pathophysiology.


Asunto(s)
Electroencefalografía , Síndrome del Cromosoma X Frágil/fisiopatología , Estimulación Acústica , Animales , Corteza Auditiva/fisiopatología , Biomarcadores , Modelos Animales de Enfermedad , Potenciales Evocados , Potenciales Evocados Auditivos , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Ratones , Ratones Noqueados , Microelectrodos , Fenotipo
5.
Cereb Cortex ; 30(3): 969-988, 2020 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-31364704

RESUMEN

Fragile X syndrome (FXS) is a leading genetic cause of autism with symptoms that include sensory processing deficits. In both humans with FXS and a mouse model [Fmr1 knockout (KO) mouse], electroencephalographic (EEG) recordings show enhanced resting state gamma power and reduced sound-evoked gamma synchrony. We previously showed that elevated levels of matrix metalloproteinase-9 (MMP-9) may contribute to these phenotypes by affecting perineuronal nets (PNNs) around parvalbumin (PV) interneurons in the auditory cortex of Fmr1 KO mice. However, how different cell types within local cortical circuits contribute to these deficits is not known. Here, we examined whether Fmr1 deletion in forebrain excitatory neurons affects neural oscillations, MMP-9 activity, and PV/PNN expression in the auditory cortex. We found that cortical MMP-9 gelatinase activity, mTOR/Akt phosphorylation, and resting EEG gamma power were enhanced in CreNex1/Fmr1Flox/y conditional KO (cKO) mice, whereas the density of PV/PNN cells was reduced. The CreNex1/Fmr1Flox/y cKO mice also show increased locomotor activity, but not the anxiety-like behaviors. These results indicate that fragile X mental retardation protein changes in excitatory neurons in the cortex are sufficient to elicit cellular, electrophysiological, and behavioral phenotypes in Fmr1 KO mice. More broadly, these results indicate that local cortical circuit abnormalities contribute to sensory processing deficits in autism spectrum disorders.


Asunto(s)
Corteza Auditiva/fisiopatología , Conducta Animal , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/fisiología , Síndrome del Cromosoma X Frágil/fisiopatología , Neuronas/fisiología , Prosencéfalo/fisiopatología , Estimulación Acústica , Animales , Modelos Animales de Enfermedad , Electroencefalografía , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Ritmo Gamma , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Transducción de Señal
6.
Brain Res ; 1719: 24-29, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31128097

RESUMEN

Sensory processing abnormalities are consistently associated with autism, but the underlying mechanisms and treatment options are unclear. Fragile X Syndrome (FXS) is the leading known genetic cause of intellectual disabilities and autism. One debilitating symptom of FXS is hypersensitivity to sensory stimuli. Sensory hypersensitivity is seen in both humans with FXS and FXS mouse model, the Fmr1 knock out (Fmr1 KO) mouse. Abnormal sensorimotor gating may play a role in the hypersensitivity to sensory stimuli. Humans with FXS and Fmr1 KO mice show abnormalities in acoustic startle response (ASR) and prepulse inhibition (PPI) of startle, responses commonly used to quantify sensorimotor gating. Recent studies have suggested high levels of matrix metalloproteinase-9 (MMP-9) as a potential mechanism of sensory abnormalities in FXS. Here we tested the hypothesis that genetic reduction of MMP-9 in Fmr1 KO mice rescues ASR and PPI phenotypes in adult Fmr1 KO mice. We measured MMP-9 levels in the inferior colliculus (IC), an integral region of the PPI circuit, of WT and Fmr1 KO mice at P7, P12, P18, and P40. MMP-9 levels were higher in the IC of Fmr1 KO mice during early development (P7, P12), but not in adults. We compared ASR and PPI responses in young (P23-25) and adult (P50-80) Fmr1 KO mice to their age-matched wildtype (WT) controls. We found that both ASR and PPI were reduced in the young Fmr1 KO mice compared to age-matched WT mice. There was no genotype difference for ASR in the adult mice, but PPI was significantly reduced in the adult Fmr1 KO mice. The adult mouse data are similar to those observed in humans with FXS. Genetic reduction of MMP-9 in the Fmr1 KO mice resulted in a rescue of adult PPI responses to WT levels. Taken together, these results show sensorimotor gating abnormalities in Fmr1 KO mice, and suggest the potential for MMP-9 regulation as a therapeutic target to reduce sensory hypersensitivity.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Metaloproteinasa 9 de la Matriz/genética , Inhibición Prepulso/fisiología , Reflejo de Sobresalto/genética , Estimulación Acústica/métodos , Animales , Modelos Animales de Enfermedad , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/fisiopatología , Genotipo , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Fenotipo , Inhibición Prepulso/genética , Filtrado Sensorial/genética
7.
Neurobiol Dis ; 115: 39-48, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29605426

RESUMEN

Identification of comparable biomarkers in humans and validated animal models will facilitate pre-clinical to clinical therapeutic pipelines to treat neurodevelopmental disorders. Fragile X Syndrome (FXS) is a leading known genetic cause of intellectual disability with symptoms that include increased anxiety, social and sensory processing deficits. Recent EEG studies in humans with FXS have identified neural oscillation deficits that include enhanced resting state gamma power and reduced inter-trial coherence of sound evoked gamma oscillations. To determine if analogous phenotypes are present in an animal model of FXS, we recorded EEGs in awake, freely moving Fmr1 knock out (KO) mice using similar stimuli as in the human studies. We report remarkably similar neural oscillation phenotypes in the Fmr1 KO mouse including enhanced resting state gamma power and reduced evoked gamma synchronization. The gamma band inter-trial coherence of neural response was reduced in both auditory and frontal cortex of Fmr1 KO mice stimulated with a sound whose envelope was modulated from 1 to 100 Hz, similar to that seen in humans with FXS. These deficits suggest a form of enhanced 'resting state noise' that interferes with the ability of the circuit to mount a synchronized response to sensory input, predicting specific sensory and cognitive deficits in FXS. The abnormal gamma oscillations are consistent with parvalbumin neuron and perineuronal net deficits seen in the Fmr1 KO mouse auditory cortex indicating that the EEG biomarkers are not only clinically relevant, but could also be used to probe cellular and circuit mechanisms of sensory hypersensitivity in FXS.


Asunto(s)
Modelos Animales de Enfermedad , Electroencefalografía/métodos , Síndrome del Cromosoma X Frágil/fisiopatología , Fenotipo , Investigación Biomédica Traslacional/métodos , Estimulación Acústica/métodos , Animales , Corteza Auditiva/fisiopatología , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Lóbulo Frontal/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
8.
Stereotact Funct Neurosurg ; 80(1-4): 28-31, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14745205

RESUMEN

BACKGROUND: The incidence of intracranial hemorrhage occurring during microelectrode-guided implantation of deep brain stimulators (DBS) for movement disorders has not been well defined. We report the incidence of hemorrhage in a large series of DBS implants into the subthalamic nucleus (STN), thalamus (VIM) and internal globus pallidus (GPi). METHODS: All DBS procedures performed by a single surgeon (P.A.S.) between June 1998 and April 2003 were included in this study. Patients had postoperative imaging (MRI or CT) 4-24 h following surgery, and all hematomas >0.2 cm(3) in volume were noted and scored as symptomatic (associated with any new neurologic deficit lasting >24 h) or asymptomatic. RESULTS: The total number of lead implants was 357. There were 5 symptomatic hematomas and 6 asymptomatic hematomas. The relative risk of hematoma (any type) per lead implant was 3.1%. The incidence of hematoma by target site was 2.5% per lead for STN-DBS, 6.7% for GPi-DBS and 0% for VIM-DBS. CONCLUSION: The overall risk of intraoperative or early postoperative symptomatic hemorrhage with microelectrode-guided DBS, over all targets, was 1.4% per lead implant. The brain target had a significant effect on the risk of hemorrhage.


Asunto(s)
Hemorragia Cerebral/etiología , Terapia por Estimulación Eléctrica/efectos adversos , Microelectrodos/efectos adversos , Trastornos del Movimiento/terapia , Hemorragia Cerebral/epidemiología , Globo Pálido/cirugía , Humanos , Incidencia , Trastornos del Movimiento/epidemiología , Trastornos del Movimiento/cirugía , Complicaciones Posoperatorias/epidemiología , Factores de Riesgo , Núcleo Subtalámico/cirugía , Tálamo/cirugía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA