Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Physiol ; 599(23): 5261-5279, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34676533

RESUMEN

Vagus nerve stimulation (VNS) treats patients with drug-resistant epilepsy, depression and heart failure, but the mechanisms responsible are uncertain. The mild stimulus intensities used in chronic VNS suggest activation of myelinated primary visceral afferents projecting to the nucleus of the solitary tract (NTS). Here, we monitored the activity of second and higher order NTS neurons in response to peripheral vagal activation using therapeutic VNS criteria. A bipolar stimulating electrode activated the left cervical vagus nerve, and stereotaxically placed single tungsten electrodes recorded unit activity from the left caudomedial NTS of chloralose-anaesthetized rats. High-intensity single electrical stimuli established vagal afferent conduction velocity (myelinated A-type or unmyelinated C-type) as well as synaptic order (second vs. higher order using paired electrical stimuli) for inputs to single NTS neurons. Then, VNS treatment was applied. A mid-collicular knife cut (KC) divided the brainstem from all supramedullary regions to determine their contribution to NTS activity. Our chief findings indicate that the KC reduced basal spontaneous activity of second-order NTS neurons receiving myelinated vagal input by 85%. In these neurons, acute VNS increased activity similarly in Control and KC animals. Interestingly, the KC interrupted VNS activation of higher order NTS neurons and second-order NTS neurons receiving unmyelinated vagal input, indicating that supramedullary descending projections to NTS are needed to amplify the peripheral neuronal signal from VNS. The present study begins to define the pathways activated during VNS and will help to better identify the central nervous system contributions to the therapeutic benefits of VNS therapy. KEY POINTS: Vagus nerve stimulation is routinely used in the clinic to treat epilepsy and depression, despite our uncertainty about how this treatment works. For this study, the connections between the nucleus of the solitary tract (NTS) and the higher brain regions were severed to learn more about their contribution to activity of these neurons during stimulation. Severing these brain connections reduced baseline activity as well as reducing stimulation-induced activation for NTS neurons receiving myelinated vagal input. Higher brain regions play a significant role in maintaining both normal activity in NTS and indirect mechanisms of enhancing NTS neuronal activity during vagus nerve stimulation.


Asunto(s)
Estimulación del Nervio Vago , Animales , Tronco Encefálico , Estimulación Eléctrica , Humanos , Neuronas , Ratas , Núcleo Solitario , Nervio Vago
2.
Brain Stimul ; 13(5): 1323-1332, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32629028

RESUMEN

BACKGROUND: Vagus nerve stimulation (VNS) modifies brain rhythms in the locus coeruleus (LC) via the solitary nucleus. Degeneration of the LC in Parkinson's disease (PD) is an early catalyst of the spreading neurodegenerative process, suggesting that stimulating LC output with VNS has the potential to modify disease progression. We previously showed in a lesion PD model that VNS delivered twice daily reduced neuroinflammation and motor deficits, and attenuated tyrosine hydroxylase (TH)-positive cell loss. OBJECTIVE: The goal of this study was to characterize the differential effects of three clinically-relevant VNS paradigms in a PD lesion model. METHODS: Eleven days after DSP-4 (N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine, noradrenergic lesion, administered systemically)/6-OHDA (6-hydroxydopamine, dopaminergic lesion, administered intrastriatally) rats were implanted with VNS devices, and received either low-frequency VNS, standard-frequency VNS, or high-frequency microburst VNS. After 10 days of treatment and behavioral assessment, rats were euthanized, right prefrontal cortex (PFC) was dissected for norepinephrine assessment, and the left striatum, bilateral substantia nigra (SN), and LC were sectioned for immunohistochemical detection of catecholamine neurons, α-synuclein, astrocytes, and microglia. RESULTS: At higher VNS frequencies, specifically microburst VNS, greater improvements occurred in motor function, attenuation of TH-positive cell loss in SN and LC, and norepinephrine concentration in the PFC. Additionally, higher VNS frequencies resulted in lower intrasomal α-synuclein accumulation and glial density in the SN. CONCLUSIONS: These data indicate that higher stimulation frequencies provided the greatest attenuation of behavioral and pathological markers in this PD model, indicating therapeutic potential for these VNS paradigms.


Asunto(s)
Locomoción/fisiología , Locus Coeruleus/metabolismo , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/terapia , Sustancia Negra/metabolismo , Estimulación del Nervio Vago/métodos , Animales , Locomoción/efectos de los fármacos , Locus Coeruleus/efectos de los fármacos , Masculino , Oxidopamina/toxicidad , Trastornos Parkinsonianos/inducido químicamente , Ratas , Ratas Long-Evans , Sustancia Negra/efectos de los fármacos , Tirosina 3-Monooxigenasa/metabolismo
3.
Exp Neurol ; 330: 113315, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32302678

RESUMEN

Parkinson's disease (PD), a debilitating progressive degenerative movement disorder associated with loss of dopaminergic (DA) neurons in the substantia nigra (SN), afflicts approximately one million people in the U.S., including a significant number of Veterans. Disease characteristics include tremor, rigidity, postural instability, bradykinesia, and at a cellular level, glial cell activation and Lewy body inclusions in DA neurons. The most potent medical/surgical treatments do not ultimately prevent disease progression. Therefore, new therapies must be developed to halt progression of the disease. While the mechanisms of the degenerative process in PD remain elusive, chronic inflammation, a common factor in many neurodegenerative diseases, has been implicated with associated accumulation of toxic aggregated α-synuclein in neurons. Calpain, a calcium-activated cysteine neutral protease, plays a pivotal role in SN and spinal cord degeneration in PD via its role in α-synuclein aggregation, activation/migration of microglia and T cells, and upregulation of inflammatory processes. Here we report an increased expression of a subset of CD4+ T cells in rodent models of PD, including MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) mice and DSP-4 [N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine hydrochloride]/6-hydroxydopamine rats, which produced higher levels of perforin and granzyme B - typically found in cytotoxic T cells. Importantly, the CD4+ cytotoxic subtype was attenuated following calpain inhibition in MPTP mice, suggesting that calpain and this distinct CD4+ T cell subset may have critical roles in the inflammatory process, disease progression, and neurodegeneration in PD.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Calpaína/inmunología , Trastornos Parkinsonianos/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Calpaína/metabolismo , Modelos Animales de Enfermedad , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Trastornos Parkinsonianos/patología , Ratas , Ratas Long-Evans , Subgrupos de Linfocitos T/inmunología
4.
Behav Brain Res ; 373: 112080, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31301412

RESUMEN

Vagus nerve stimulation (VNS) is being explored as a potential therapeutic for Parkinson's disease (PD). VNS is less invasive than other surgical treatments and has beneficial effects on behavior and brain pathology. It has been suggested that VNS exerts these effects by increasing brain-derived neurotrophic factor (BDNF) to enhance pro-survival mechanisms of its receptor, tropomyosin receptor kinase-B (TrkB). We have previously shown that striatal BDNF is increased after VNS in a lesion model of PD. By chronically administering ANA-12, a TrkB-specific antagonist, we aimed to determine TrkB's role in beneficial VNS effects for a PD model. In this study, we administered a noradrenergic neurotoxin, DSP-4, intraperitoneally and one week later administered a bilateral intrastriatal dopaminergic neurotoxin, 6-OHDA. At this time, the left vagus nerve was cuffed for stimulation. Eleven days later, rats received VNS twice per day for ten days, with daily locomotor assessment. Daily ANA-12 injections were given one hour prior to the afternoon stimulation and concurrent locomotor session. Following the final VNS session, rats were euthanized, and left striatum, bilateral substantia nigra and locus coeruleus were sectioned for immunohistochemical detection of neurons, α-synuclein, astrocytes, and microglia. While ANA-12 did not avert behavioral improvements of VNS, and only partially prevented VNS-induced attenuation of neuronal loss in the locus coeruleus, it did stop neuronal and anti-inflammatory effects of VNS in the nigrostriatal system, indicating a role for TrkB in mediating VNS efficacy. However, our data also suggest that BDNF-TrkB is not the sole mechanism of action for VNS in PD.


Asunto(s)
Enfermedad de Parkinson/metabolismo , Receptor trkB/metabolismo , Nervio Vago/metabolismo , Animales , Azepinas/farmacología , Benzamidas/farmacología , Encéfalo/metabolismo , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Dopamina/farmacología , Locus Coeruleus/metabolismo , Masculino , Neostriado/metabolismo , Norepinefrina/farmacología , Oxidopamina/farmacología , Enfermedad de Parkinson/patología , Ratas , Ratas Long-Evans , Receptor trkB/fisiología , Sustancia Negra/metabolismo , Estimulación del Nervio Vago/métodos
5.
Brain Stimul ; 10(6): 1045-1054, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28918943

RESUMEN

BACKGROUND: Parkinson's disease (PD) is a progressive, neurodegenerative disorder with no disease-modifying therapies, and symptomatic treatments are often limited by debilitating side effects. In PD, locus coeruleus noradrenergic (LC-NE) neurons degenerate prior to substantia nigra dopaminergic (SN-DA) neurons. Vagus nerve stimulation (VNS) activates LC neurons, and decreases pro-inflammatory markers, allowing improvement of LC targets, making it a potential PD therapeutic. OBJECTIVE: To assess therapeutic potential of VNS in a PD model. METHODS: To mimic the progression of PD degeneration, rats received a systemic injection of noradrenergic neurotoxin DSP-4, followed one week later by bilateral intrastriatal injection of dopaminergic neurotoxin 6-hydroxydopamine. At this time, a subset of rats also had vagus cuffs implanted. After eleven days, rats received a precise VNS regimen twice a day for ten days, and locomotion was measured during each afternoon session. Immediately following final stimulation, rats were euthanized, and left dorsal striatum, bilateral SN and LC were sectioned for immunohistochemical detection of monoaminergic neurons (tyrosine hydroxylase, TH), α-synuclein, astrocytes (GFAP) and microglia (Iba-1). RESULTS: VNS significantly increased locomotion of lesioned rats. VNS also resulted in increased expression of TH in striatum, SN, and LC; decreased SN α-synuclein expression; and decreased expression of glial markers in the SN and LC of lesioned rats. Additionally, saline-treated rats after VNS, had higher LC TH and lower SN Iba-1. CONCLUSIONS: Our findings of increased locomotion, beneficial effects on LC-NE and SN-DA neurons, decreased α-synuclein density in SN TH-positive neurons, and neuroinflammation suggest VNS has potential as a novel PD therapeutic.


Asunto(s)
Neuronas Adrenérgicas/metabolismo , Neuronas Dopaminérgicas/metabolismo , Locomoción/fisiología , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/terapia , Estimulación del Nervio Vago/métodos , Neuronas Adrenérgicas/efectos de los fármacos , Animales , Bencilaminas/toxicidad , Dopamina/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Locomoción/efectos de los fármacos , Locus Coeruleus/efectos de los fármacos , Locus Coeruleus/metabolismo , Masculino , Neostriado/efectos de los fármacos , Neostriado/metabolismo , Norepinefrina/metabolismo , Oxidopamina/toxicidad , Trastornos Parkinsonianos/inducido químicamente , Distribución Aleatoria , Ratas , Ratas Long-Evans , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
6.
Brain Res ; 1651: 88-94, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27659966

RESUMEN

Mitochondrial dysfunction has been implicated in the degeneration of dopamine (DA) neurons in Parkinson's disease (PD). In addition, animal models of PD utilizing neurotoxins, such as 6-hydroxydopamine and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, have shown that these toxins disrupt mitochondrial respiration by targeting complex I of the electron transport chain, thereby impairing DA neurons in these models. A MitoPark mouse model was created to mimic the mitochondrial dysfunction observed in the DA system of PD patients. These mice display the same phenotypic characteristics as PD, including accelerated decline in motor function and DAergic systems with age. Previously, these mice have responded to L-Dopa treatment and develop L-Dopa induced dyskinesia (LID) as they age. A potential mechanism involved in the formation of LID is greater glutamate release into the dorsal striatum as a result of altered basal ganglia neurocircuitry due to reduced nigrostriatal DA neurotransmission. Therefore, the focus of this study was to assess various indicators of glutamate neurotransmission in the dorsal striatum of MitoPark mice at an age in which nigrostriatal DA has degenerated. At 28 weeks of age, MitoPark mice had, upon KCl stimulation, greater glutamate release in the dorsal striatum compared to control mice. In addition, uptake kinetics were slower in MitoPark mice. These findings were coupled with reduced expression of the glutamate re-uptake transporter, GLT-1, thus providing an environment suitable for glutamate excitotoxic events, leading to altered physiological function in these mice.


Asunto(s)
Cuerpo Estriado/metabolismo , Ácido Glutámico/metabolismo , Mitocondrias/metabolismo , Trastornos Parkinsonianos/metabolismo , Transmisión Sináptica/fisiología , Animales , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/patología , Dopamina/metabolismo , Transportador 2 de Aminoácidos Excitadores/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Cinética , Masculino , Ratones Transgénicos , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Neurotransmisores/farmacología , Trastornos Parkinsonianos/patología , Cloruro de Potasio/farmacología , Receptor del Glutamato Metabotropico 5/metabolismo , Transmisión Sináptica/efectos de los fármacos
7.
Neurobiol Aging ; 36(3): 1569-76, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25577412

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) helps protect dopaminergic neurons in the nigrostriatal tract. Although the cause of nigrostriatal degeneration is unknown, one theory is that excess glutamate from the subthalamic nucleus results in excitotoxic events in the substantia nigra (SN). Because dopaminergic degeneration is accompanied by a reduction in GDNF, we examined glutamate neurotransmission in the SN using a Gdnf heterozygous mouse model (Gdnf(+/-)) at 8 and 12 months of age. At 8 months, Gdnf(+/-) mice have greater glutamate release and higher basal glutamate levels, which precede the SN dopaminergic degeneration observed at 12 months of age. However, at 12 months, Gdnf(+/-) mice have lower basal levels of glutamate and less glutamate release than wild-type mice. Also at 8 months, Gdnf(+/-) mice have lower levels of glutamate transporter-1 and greater glial fibrillary acidic protein levels in the SN compared with wild-type mice, differences that increase with age. These data suggest that reduced levels of GDNF induce excess glutamate release and dysregulation of glutamate transporter-1, causing excitotoxicity in the SN that precedes dopaminergic degeneration.


Asunto(s)
Envejecimiento/fisiología , Transportador 2 de Aminoácidos Excitadores/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/deficiencia , Factor Neurotrófico Derivado de la Línea Celular Glial/fisiología , Ácido Glutámico/metabolismo , Degeneración Nerviosa/etiología , Degeneración Nerviosa/patología , Sustancia Negra/patología , Sustancia Negra/fisiopatología , Transmisión Sináptica/genética , Transmisión Sináptica/fisiología , Envejecimiento/genética , Envejecimiento/patología , Animales , Neuronas Dopaminérgicas/patología , Femenino , Heterocigoto , Masculino , Ratones Endogámicos C57BL , Núcleo Subtalámico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA