Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Ann Plast Surg ; 90(3): 237-241, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36796045

RESUMEN

BACKGROUND: Electrical stimulation can accelerate peripheral nerve regeneration after injury and repair. Clinically, direct electrical stimulation (DES) may involve longer operating times, increasing risks of perioperative complications. Transcutaneous electrical stimulation (TCES) is a noninvasive alternative. In this study, we investigate how transcutaneous and DES compare for accelerating functional nerve recovery in a mouse sciatic nerve model. METHODS: Twenty-eight mice were divided into sham (n = 4), axotomy (n = 8), DES (n = 8), and TCES (n = 8) groups. After sciatic nerve transection and repair, the proximal nerve was subjected to DES or TCES at 20 Hz for 1 hour. Sciatic functional index was measured before the injury, and at weeks 1, 2, 4, 6, 8, 10, and 12 by walking-track analysis. Electrophysiological measures were taken at week 12. RESULTS: Kinematic studies showed significant improvement from the 8th week to the 12th week for both electrical stimulation groups compared with the axotomy group (P < 0.05), with no difference between the electrical stimulation groups. At the 12th week, both DES and TCES groups had significantly faster average conduction velocity than the axotomy group. CONCLUSIONS: Functional recovery was significantly better from 8 weeks onward in mice receiving either DES or TCES stimulation when compared with axotomy and repair alone. Transcutaneous electrical stimulation is a minimally invasive alternative treatment for accelerating functional recovery after peripheral nerve injury.


Asunto(s)
Traumatismos de los Nervios Periféricos , Nervio Ciático , Ratones , Animales , Nervio Ciático/cirugía , Nervio Ciático/lesiones , Traumatismos de los Nervios Periféricos/cirugía , Axotomía , Regeneración Nerviosa/fisiología , Recuperación de la Función/fisiología , Estimulación Eléctrica
2.
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
3.
J Physiol ; 595(22): 6887-6903, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28862330

RESUMEN

KEY POINTS: The evoked cardiac response to bipolar cervical vagus nerve stimulation (VNS) reflects a dynamic interaction between afferent mediated decreases in central parasympathetic drive and suppressive effects evoked by direct stimulation of parasympathetic efferent axons to the heart. The neural fulcrum is defined as the operating point, based on frequency-amplitude-pulse width, where a null heart rate response is reproducibly evoked during the on-phase of VNS. Cardiac control, based on the principal of the neural fulcrum, can be elicited from either vagus. Beta-receptor blockade does not alter the tachycardia phase to low intensity VNS, but can increase the bradycardia to higher intensity VNS. While muscarinic cholinergic blockade prevented the VNS-induced bradycardia, clinically relevant doses of ACE inhibitors, beta-blockade and the funny channel blocker ivabradine did not alter the VNS chronotropic response. While there are qualitative differences in VNS heart control between awake and anaesthetized states, the physiological expression of the neural fulcrum is maintained. ABSTRACT: Vagus nerve stimulation (VNS) is an emerging therapy for treatment of chronic heart failure and remains a standard of therapy in patients with treatment-resistant epilepsy. The objective of this work was to characterize heart rate (HR) responses (HRRs) during the active phase of chronic VNS over a wide range of stimulation parameters in order to define optimal protocols for bidirectional bioelectronic control of the heart. In normal canines, bipolar electrodes were chronically implanted on the cervical vagosympathetic trunk bilaterally with anode cephalad to cathode (n = 8, 'cardiac' configuration) or with electrode positions reversed (n = 8, 'epilepsy' configuration). In awake state, HRRs were determined for each combination of pulse frequency (2-20 Hz), intensity (0-3.5 mA) and pulse widths (130-750 µs) over 14 months. At low intensities and higher frequency VNS, HR increased during the VNS active phase owing to afferent modulation of parasympathetic central drive. When functional effects of afferent and efferent fibre activation were balanced, a null HRR was evoked (defined as 'neural fulcrum') during which HRR ≈ 0. As intensity increased further, HR was reduced during the active phase of VNS. While qualitatively similar, VNS delivered in the epilepsy configuration resulted in more pronounced HR acceleration and reduced HR deceleration during VNS. At termination, under anaesthesia, transection of the vagi rostral to the stimulation site eliminated the augmenting response to VNS and enhanced the parasympathetic efferent-mediated suppressing effect on electrical and mechanical function of the heart. In conclusion, VNS activates central then peripheral aspects of the cardiac nervous system. VNS control over cardiac function is maintained during chronic therapy.


Asunto(s)
Frecuencia Cardíaca , Corazón/fisiología , Estimulación del Nervio Vago , Nervio Vago/fisiología , Antagonistas Adrenérgicos beta/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Benzazepinas/farmacología , Perros , Femenino , Corazón/inervación , Ivabradina , Masculino , Antagonistas Muscarínicos/farmacología , Nervio Vago/efectos de los fármacos
4.
Am J Physiol Heart Circ Physiol ; 313(2): H354-H367, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28476920

RESUMEN

Vagus nerve stimulation (VNS) currently treats patients with drug-resistant epilepsy, depression, and heart failure. The mild intensities used in chronic VNS suggest that primary visceral afferents and central nervous system activation are involved. Here, we measured the activity of neurons in the nucleus of the solitary tract (NTS) in anesthetized rats using clinically styled VNS. Our chief findings indicate that VNS at threshold bradycardic intensity activated NTS neuron discharge in one-third of NTS neurons. This VNS directly activated only myelinated vagal afferents projecting to second-order NTS neurons. Most VNS-induced activity in NTS, however, was unsynchronized to vagal stimuli. Thus, VNS activated unsynchronized activity in NTS neurons that were second order to vagal afferent C-fibers as well as higher-order NTS neurons only polysynaptically activated by the vagus. Overall, cardiovascular-sensitive and -insensitive NTS neurons were similarly activated by VNS: 3/4 neurons with monosynaptic vagal A-fiber afferents, 6/42 neurons with monosynaptic vagal C-fiber afferents, and 16/21 polysynaptic NTS neurons. Provocatively, vagal A-fibers indirectly activated C-fiber neurons during VNS. Elevated spontaneous spiking was quantitatively much higher than synchronized activity and extended well into the periods of nonstimulation. Surprisingly, many polysynaptic NTS neurons responded to half the bradycardic intensity used in clinical studies, indicating that a subset of myelinated vagal afferents is sufficient to evoke VNS indirect activation. Our study uncovered a myelinated vagal afferent drive that indirectly activates NTS neurons and thus central pathways beyond NTS and support reconsideration of brain contributions of vagal afferents underpinning of therapeutic impacts.NEW & NOTEWORTHY Acute vagus nerve stimulation elevated activity in neurons located in the medial nucleus of the solitary tract. Such stimuli directly activated only myelinated vagal afferents but indirectly activated a subpopulation of second- and higher-order neurons, suggesting that afferent mechanisms and central neuron activation may be responsible for vagus nerve stimulation efficacy.


Asunto(s)
Potenciales de Acción , Potenciales Evocados , Fibras Nerviosas Mielínicas/fisiología , Fibras Nerviosas Amielínicas/fisiología , Células Receptoras Sensoriales/fisiología , Núcleo Solitario/fisiología , Estimulación del Nervio Vago/métodos , Nervio Vago/fisiología , Animales , Barorreflejo , Presión Sanguínea , Bradicardia/etiología , Bradicardia/fisiopatología , Frecuencia Cardíaca , Masculino , Modelos Animales , Vías Nerviosas/fisiología , Ratas Sprague-Dawley , Estimulación del Nervio Vago/efectos adversos
5.
Am J Physiol Gastrointest Liver Physiol ; 311(4): G754-G762, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27562060

RESUMEN

Altered gut microbial diversity has been associated with several chronic disease states, including heart failure. Stimulation of the vagus nerve, which innervates the heart and abdominal organs, is proving to be an effective therapeutic in heart failure. We hypothesized that cervical vagus nerve stimulation (VNS) could alter fecal flora and prevent aberrations observed in fecal samples from heart failure animals. To determine whether microbial abundances were altered by pressure overload (PO), leading to heart failure and VNS therapy, a VNS pulse generator was implanted with a stimulus lead on either the left or right vagus nerve before creation of PO by aortic constriction. Animals received intermittent, open-loop stimulation or sham treatment, and their heart function was monitored by echocardiography. Left ventricular end-systolic and diastolic volumes, as well as cardiac output, were impaired in PO animals compared with baseline. VNS mitigated these effects. Metagenetic analysis was then performed using 16S rRNA sequencing to identify bacterial genera present in fecal samples. The abundance of 10 genera was significantly altered by PO, 8 of which were mitigated in animals receiving either left- or right-sided VNS. Metatranscriptomics analyses indicate that the abundance of genera that express genes associated with ATP-binding cassette transport and amino sugar/nitrogen metabolism was significantly changed following PO. These gut flora changes were not observed in PO animals subjected to VNS. These data suggest that VNS prevents aberrant gut flora following PO, which could contribute to its beneficial effects in heart failure patients.


Asunto(s)
Heces/microbiología , Corazón/fisiopatología , Estimulación del Nervio Vago , Disfunción Ventricular Izquierda/terapia , Animales , Cobayas , Masculino , Disfunción Ventricular Izquierda/microbiología , Disfunción Ventricular Izquierda/fisiopatología
6.
Am J Physiol Heart Circ Physiol ; 311(5): H1311-H1320, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27591222

RESUMEN

Mediastinal nerve stimulation (MNS) reproducibly evokes atrial fibrillation (AF) by excessive and heterogeneous activation of intrinsic cardiac (IC) neurons. This study evaluated whether preemptive vagus nerve stimulation (VNS) impacts MNS-induced evoked changes in IC neural network activity to thereby alter susceptibility to AF. IC neuronal activity in the right atrial ganglionated plexus was directly recorded in anesthetized canines (n = 8) using a linear microelectrode array concomitant with right atrial electrical activity in response to: 1) epicardial touch or great vessel occlusion vs. 2) stellate or vagal stimulation. From these stressors, post hoc analysis (based on the Skellam distribution) defined IC neurons so recorded as afferent, efferent, or convergent (afferent and efferent inputs) local circuit neurons (LCN). The capacity of right-sided MNS to modify IC activity in the induction of AF was determined before and after preemptive right (RCV)- vs. left (LCV)-sided VNS (15 Hz, 500 µs; 1.2× bradycardia threshold). Neuronal (n = 89) activity at baseline (0.11 ± 0.29 Hz) increased during MNS-induced AF (0.51 ± 1.30 Hz; P < 0.001). Convergent LCNs were preferentially activated by MNS. Preemptive RCV reduced MNS-induced changes in LCN activity (by 70%) while mitigating MNS-induced AF (by 75%). Preemptive LCV reduced LCN activity by 60% while mitigating AF potential by 40%. IC neuronal synchrony increased during neurally induced AF, a local neural network response mitigated by preemptive VNS. These antiarrhythmic effects persisted post-VNS for, on average, 26 min. In conclusion, VNS preferentially targets convergent LCNs and their interactive coherence to mitigate the potential for neurally induced AF. The antiarrhythmic properties imposed by VNS exhibit memory.


Asunto(s)
Fibrilación Atrial/fisiopatología , Atrios Cardíacos/inervación , Miocardio/citología , Neuronas/fisiología , Estimulación del Nervio Vago , Animales , Perros , Mediastino/inervación , Red Nerviosa , Nervio Vago
7.
Am J Physiol Heart Circ Physiol ; 310(10): H1349-59, 2016 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-26993230

RESUMEN

Our objective was to determine whether chronic vagus nerve stimulation (VNS) mitigates pressure overload (PO)-induced remodeling of the cardioneural interface. Guinea pigs (n = 48) were randomized to right or left cervical vagus (RCV or LCV) implant. After 2 wk, chronic left ventricular PO was induced by partial (15-20%) aortic constriction. Of the 31 animals surviving PO induction, 10 were randomized to RCV VNS, 9 to LCV VNS, and 12 to sham VNS. VNS was delivered at 20 Hz and 1.14 ± 0.03 mA at a 22% duty cycle. VNS commenced 10 days after PO induction and was maintained for 40 days. Time-matched controls (n = 9) were evaluated concurrently. Echocardiograms were obtained before and 50 days after PO. At termination, intracellular current-clamp recordings of intrinsic cardiac (IC) neurons were studied in vitro to determine effects of therapy on soma characteristics. Ventricular cardiomyocyte sizes were assessed with histology along with immunoblot analysis of selected proteins in myocardial tissue extracts. In sham-treated animals, PO increased cardiac output (34%, P < 0.004), as well as systolic (114%, P < 0.04) and diastolic (49%, P < 0.002) left ventricular volumes, a hemodynamic response prevented by VNS. PO-induced enhancements of IC synaptic efficacy and muscarinic sensitivity of IC neurons were mitigated by chronic VNS. Increased myocyte size, which doubled in PO (P < 0.05), was mitigated by RCV. PO hypertrophic myocardium displayed decreased glycogen synthase (GS) protein levels and accumulation of the phosphorylated (inactive) form of GS. These PO-induced changes in GS were moderated by left VNS. Chronic VNS targets IC neurons accompanying PO to obtund associated adverse cardiomyocyte remodeling.


Asunto(s)
Corazón/inervación , Hipertrofia Ventricular Izquierda/terapia , Estimulación del Nervio Vago , Nervio Vago/fisiopatología , Función Ventricular Izquierda , Presión Ventricular , Remodelación Ventricular , Animales , Apoptosis , Modelos Animales de Enfermedad , Glucógeno Sintasa/metabolismo , Cobayas , Hipertrofia Ventricular Izquierda/etiología , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/fisiopatología , Masculino , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fosforilación , Transmisión Sináptica , Factores de Tiempo
8.
Proc Natl Acad Sci U S A ; 110(24): 9974-9, 2013 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-23716671

RESUMEN

Maintaining wakefulness is associated with a progressive increase in the need for sleep. This phenomenon has been linked to changes in synaptic function. The synaptic adhesion molecule Neuroligin-1 (NLG1) controls the activity and synaptic localization of N-methyl-d-aspartate receptors, which activity is impaired by prolonged wakefulness. We here highlight that this pathway may underlie both the adverse effects of sleep loss on cognition and the subsequent changes in cortical synchrony. We found that the expression of specific Nlg1 transcript variants is changed by sleep deprivation in three mouse strains. These observations were associated with strain-specific changes in synaptic NLG1 protein content. Importantly, we showed that Nlg1 knockout mice are not able to sustain wakefulness and spend more time in nonrapid eye movement sleep than wild-type mice. These changes occurred with modifications in waking quality as exemplified by low theta/alpha activity during wakefulness and poor preference for social novelty, as well as altered delta synchrony during sleep. Finally, we identified a transcriptional pathway that could underlie the sleep/wake-dependent changes in Nlg1 expression and that involves clock transcription factors. We thus suggest that NLG1 is an element that contributes to the coupling of neuronal activity to sleep/wake regulation.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/fisiología , Neuronas/fisiología , Sueño/fisiología , Vigilia/fisiología , Animales , Western Blotting , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Electroencefalografía , Electromiografía , Expresión Génica , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos AKR , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Neuronas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sueño/genética , Privación de Sueño/genética , Privación de Sueño/fisiopatología , Especificidad de la Especie , Factores de Tiempo , Vigilia/genética
9.
J Neurosci ; 34(29): 9768-78, 2014 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-25031414

RESUMEN

Paranodal axoglial junctions are critical for maintaining the segregation of axonal domains along myelinated axons; however, the proteins required to organize and maintain this structure are not fully understood. Netrin-1 and its receptor Deleted in Colorectal Cancer (DCC) are proteins enriched at paranodes that are expressed by neurons and oligodendrocytes. To identify the specific function of DCC expressed by oligodendrocytes in vivo, we selectively eliminated DCC from mature myelinating oligodendrocytes using an inducible cre regulated by the proteolipid protein promoter. We demonstrate that DCC deletion results in progressive disruption of the organization of axonal domains, myelin ultrastructure, and myelin protein composition. Conditional DCC knock-out mice develop balance and coordination deficits and exhibit decreased conduction velocity. We conclude that DCC expression by oligodendrocytes is required for the maintenance and stability of myelin in vivo, which is essential for proper signal conduction in the CNS.


Asunto(s)
Uniones Comunicantes/fisiología , Regulación del Desarrollo de la Expresión Génica , Vaina de Mielina/fisiología , Oligodendroglía/metabolismo , Receptores de Superficie Celular/deficiencia , Proteínas Supresoras de Tumor/deficiencia , Animales , Axones/fisiología , Recuento de Células , Receptor DCC , Embrión de Mamíferos , Antagonistas de Estrógenos/farmacología , Conducta Exploratoria/fisiología , Uniones Comunicantes/ultraestructura , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Integrasas/genética , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Proteína Proteolipídica de la Mielina/genética , Proteína Proteolipídica de la Mielina/metabolismo , Vaina de Mielina/ultraestructura , Conducción Nerviosa/efectos de los fármacos , Conducción Nerviosa/genética , Oligodendroglía/ultraestructura , Trastornos Psicomotores/genética , Nódulos de Ranvier/metabolismo , Nódulos de Ranvier/ultraestructura , Receptores de Superficie Celular/genética , Tamoxifeno/farmacología , Proteínas Supresoras de Tumor/genética
10.
Am J Physiol Heart Circ Physiol ; 309(7): H1198-206, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26276818

RESUMEN

This paper aims to determine whether chronic vagus nerve stimulation (VNS) mitigates myocardial infarction (MI)-induced remodeling of the intrinsic cardiac nervous system (ICNS), along with the cardiac tissue it regulates. Guinea pigs underwent VNS implantation on the right cervical vagus. Two weeks later, MI was produced by ligating the ventral descending coronary artery. VNS stimulation started 7 days post-MI (20 Hz, 0.9 ± 0.2 mA, 14 s on, 48 s off; VNS-MI, n = 7) and was compared with time-matched MI animals with sham VNS (MI n = 7) vs. untreated controls (n = 8). Echocardiograms were performed before and at 90 days post-MI. At termination, IC neuronal intracellular voltage recordings were obtained from whole-mount neuronal plexuses. MI increased left ventricular end systolic volume (LVESV) 30% (P = 0.027) and reduced LV ejection fraction (LVEF) 6.5% (P < 0.001) at 90 days post-MI compared with baseline. In the VNS-MI group, LVESV and LVEF did not differ from baseline. IC neurons showed depolarization of resting membrane potentials and increased input resistance in MI compared with VNS-MI and sham controls (P < 0.05). Neuronal excitability and sensitivity to norepinephrine increased in MI and VNS-MI groups compared with controls (P < 0.05). Synaptic efficacy, as determined by evoked responses to stimulating input axons, was reduced in VNS-MI compared with MI or controls (P < 0.05). VNS induced changes in myocytes, consistent with enhanced glycogenolysis, and blunted the MI-induced increase in the proapoptotic Bcl-2-associated X protein (P < 0.05). VNS mitigates MI-induced remodeling of the ICNS, correspondingly preserving ventricular function via both neural and cardiomyocyte-dependent actions.


Asunto(s)
Sistema Nervioso Autónomo/fisiopatología , Corazón/inervación , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/metabolismo , Plasticidad Neuronal/fisiología , Estimulación del Nervio Vago , Disfunción Ventricular Izquierda/fisiopatología , Animales , Potenciales Evocados , Glucogenólisis , Cobayas , Potenciales de la Membrana , Norepinefrina/metabolismo , Volumen Sistólico/fisiología , Transmisión Sináptica , Función Ventricular Izquierda , Proteína X Asociada a bcl-2/metabolismo
11.
J Spinal Cord Med ; 37(1): 93-100, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24090649

RESUMEN

BACKGROUND: Functional electrical stimulation (FES) has been found to be effective in restoring voluntary functions after spinal cord injury (SCI) and stroke. However, the central nervous system (CNS) changes that occur in as a result of this therapy are largely unknown. OBJECTIVE: To examine the effects of FES on the restoration of voluntary locomotor function of the CNS in a SCI rat model. METHODS: SCI rats were instrumented with chronic FES electrodes in the hindlimb muscles and were divided into two groups: (a) FES therapy and (b) sedentary. At day 7 post-SCI, the animals were assessed for locomotion performance by using a Basso, Beattie and Bresnahan (BBB) scale. They were then anesthetized for a terminal in vivo experiment. The lumbar spinal cord and somatosensory cortex were exposed and the instrumented muscles were stimulated electrically. Associated neurovascular responses in the CNS were recorded with an intrinsic optical imaging system. RESULTS: FES greatly improved locomotion recovery by day 7 post-SCI, as measured by BBB scores (P < 0.05): (a) FES 10 ± 2 and (b) controls 3 ± 1. Furthermore, the FES group showed a significant increase (P < 0.05) of neurovascular activation in the spinal cord and somatosensory cortex when the muscles were stimulated between 1 and 3 motor threshold (MT). CONCLUSION: Hind limb rehabilitation with FES is an effective strategy to improve locomotion during the acute phase post-SCI. The results of this study indicate that after FES, the CNS preserves/acquires the capacity to respond to peripheral electrical stimulation.


Asunto(s)
Vías Aferentes/fisiología , Sistema Nervioso Central/fisiopatología , Terapia por Estimulación Eléctrica/métodos , Locomoción/fisiología , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal , Animales , Modelos Animales de Enfermedad , Femenino , Lateralidad Funcional , Hemoglobinas/metabolismo , Músculo Esquelético/fisiología , Ratas , Ratas Sprague-Dawley , Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/terapia
12.
Auton Neurosci ; 253: 103162, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38513382

RESUMEN

Vagus nerve stimulation (VNS) is under clinical investigation as a therapy for heart failure with reduced ejection fraction (HFrEF). This study aimed to investigate its therapeutic effects on three main components of heart failure: cardiac function, cardiac remodeling and central neuroinflammation using a pressure overload (PO) rat model. Male Sprague-Dawley rats were divided into four groups: PO, PO + VNS, PO + VNS sham, and controls. All rats, except controls, underwent a PO surgery to constrict the thoracic aorta (~50 %) to induce HFrEF. Open loop VNS therapy was continuously administered to PO + VNS rats at 20 Hz, 1.0 mA for 60 days. Evaluation of cardiac function and structure via echocardiograms showed decreases in stroke volume and relative ejection fraction and increases in the internal diameter of the left ventricle during systole and diastole in PO rats (p < 0.05). However, these PO-induced adverse changes were alleviated with VNS therapy. Additionally, PO rats exhibited significant increases in myocyte cross sectional areas indicating hypertrophy, along with significant increases in myocardial fibrosis and apoptosis, all of which were reversed by VNS therapy (p < 0.05). Furthermore, VNS mitigated microglial activation in two central autonomic nuclei: the paraventricular nucleus of the hypothalamus and locus coeruleus. These findings demonstrate that when VNS therapy is initiated at an early stage of HFrEF progression (<10 % reduction in relative ejection fraction), the supplementation of vagal activity is effective in restoring multi organ homeostasis in a PO model.


Asunto(s)
Insuficiencia Cardíaca , Ratas Sprague-Dawley , Estimulación del Nervio Vago , Animales , Estimulación del Nervio Vago/métodos , Insuficiencia Cardíaca/terapia , Insuficiencia Cardíaca/fisiopatología , Masculino , Ratas , Modelos Animales de Enfermedad , Volumen Sistólico/fisiología , Remodelación Ventricular/fisiología , Inflamación/terapia , Inflamación/fisiopatología , Enfermedades Neuroinflamatorias/terapia , Enfermedades Neuroinflamatorias/fisiopatología
13.
J Physiol ; 591(18): 4515-33, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23818689

RESUMEN

The aims of the study were to determine how aggregates of intrinsic cardiac (IC) neurons transduce the cardiovascular milieu versus responding to changes in central neuronal drive and to determine IC network interactions subsequent to induced neural imbalances in the genesis of atrial fibrillation (AF). Activity from multiple IC neurons in the right atrial ganglionated plexus was recorded in eight anaesthetized canines using a 16-channel linear microelectrode array. Induced changes in IC neuronal activity were evaluated in response to: (1) focal cardiac mechanical distortion; (2) electrical activation of cervical vagi or stellate ganglia; (3) occlusion of the inferior vena cava or thoracic aorta; (4) transient ventricular ischaemia, and (5) neurally induced AF. Low level activity (ranging from 0 to 2.7 Hz) generated by 92 neurons was identified in basal states, activities that displayed functional interconnectivity. The majority (56%) of IC neurons so identified received indirect central inputs (vagus alone: 25%; stellate ganglion alone: 27%; both: 48%). Fifty per cent transduced the cardiac milieu responding to multimodal stressors applied to the great vessels or heart. Fifty per cent of IC neurons exhibited cardiac cycle periodicity, with activity occurring primarily in late diastole into isovolumetric contraction. Cardiac-related activity in IC neurons was primarily related to direct cardiac mechano-sensory inputs and indirect autonomic efferent inputs. In response to mediastinal nerve stimulation, most IC neurons became excessively activated; such network behaviour preceded and persisted throughout AF. It was concluded that stochastic interactions occur among IC local circuit neuronal populations in the control of regional cardiac function. Modulation of IC local circuit neuronal recruitment may represent a novel approach for the treatment of cardiac disease, including atrial arrhythmias.


Asunto(s)
Corazón/inervación , Red Nerviosa/fisiología , Neuronas/fisiología , Reflejo , Animales , Aorta Torácica/inervación , Aorta Torácica/fisiología , Fibrilación Atrial , Perros , Corazón/fisiología , Corazón/fisiopatología , Ganglio Estrellado/fisiología , Nervio Vago/fisiología , Vasoconstricción , Venas Cavas/inervación , Venas Cavas/fisiología , Disfunción Ventricular
14.
Physiol Rep ; 11(5): e15633, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36905173

RESUMEN

Vagus nerve stimulation (VNS) is used to treat drug-resistant epilepsy and depression, with additional applications under investigation. The noradrenergic center locus coeruleus (LC) is vital for VNS effects; however, the impact of varying stimulation parameters on LC activation is poorly understood. This study characterized LC activation across VNS parameters. Extracellular activity was recorded in rats' left LC while 11 VNS paradigms, utilizing variable frequencies and bursting characteristics, were pseudorandomly delivered to the left cervical vagus for five cycles. Neurons' change from baseline firing rate and timing response profiles were assessed. The proportion of neurons categorized as responders over 5 VNS cycles doubled in comparison to the first VNS cycle (p < 0.001) for all VNS paradigms, demonstrating an amplification effect. The percentage of positively consistent/positive responders increased for standard VNS paradigms with frequencies ≥10 Hz and for bursting paradigms with shorter interburst intervals and more pulses per burst. The synchrony between pairs of LC neurons increased during bursting VNS but not standard paradigms. Also, the probability of evoking a direct response during bursting VNS was higher with longer interburst intervals and a higher number of pulses per burst. Standard paradigms between 10-30 Hz best positively activates LC with consistency to VNS while the best bursting paradigm to increase activity was 300 Hz, seven pulses per burst separated by 1 s. Bursting VNS was effective in increasing synchrony between pairs of neurons, suggesting a common network recruitment originating from vagal afferents. These results indicate differential activation of LC neurons depending on the VNS parameters delivered.


Asunto(s)
Estimulación del Nervio Vago , Ratas , Animales , Estimulación del Nervio Vago/métodos , Locus Coeruleus/fisiología , Neuronas/fisiología , Norepinefrina , Nervio Vago/fisiología
15.
Am J Physiol Regul Integr Comp Physiol ; 302(3): R357-64, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22088304

RESUMEN

Our objective was to determine whether atrial fibrillation (AF) results from excessive activation of intrinsic cardiac neurons (ICNs) and, if so, whether select subpopulations of neurons therein represent therapeutic targets for suppression of this arrhythmogenic potential. Trains of five electrical stimuli (0.3-1.2 mA, 1 ms) were delivered during the atrial refractory period to mediastinal nerves (MSN) on the superior vena cava to evoke AF. Neuroanatomical studies were performed by injecting the neuronal tracer DiI into MSN sites that induced AF. Functional studies involved recording of neuronal activity in situ from the right atrial ganglionated plexus (RAGP) in response to MSN stimulation (MSNS) prior to and following neuromodulation involving either preemptive spinal cord stimulation (SCS; T(1)-T(3), 50 Hz, 200-ms duration) or ganglionic blockade (hexamethonium, 5 mg/kg). The tetramethylindocarbocyanine perchlorate (DiI) neuronal tracer labeled a subset (13.2%) of RAGP neurons, which also colocalized with cholinergic or adrenergic markers. A subset of DiI-labeled RAGP neurons were noncholinergic/nonadrenergic. MSNS evoked an ∼4-fold increase in RAGP neuronal activity from baseline, which SCS reduced by 43%. Hexamethonium blocked MSNS-evoked increases in neuronal activity. MSNS evoked AF in 78% of right-sided MSN sites, which SCS reduced to 33% and hexamethonium reduced to 7%. MSNS-induced bradycardia was maintained with SCS but was mitigated by hexamethonium. We conclude that MSNS activates subpopulations of intrinsic cardiac neurons, thereby resulting in the formation of atrial arrhythmias leading to atrial fibrillation. Stabilization of ICN local circuit neurons by SCS or the local circuit and autonomic efferent neurons with hexamethonium reduces the arrhythmogenic potential.


Asunto(s)
Fibrilación Atrial/prevención & control , Vías Autónomas/efectos de los fármacos , Bradicardia/prevención & control , Corazón/inervación , Neuronas/efectos de los fármacos , Neurotransmisores/farmacología , Animales , Fibrilación Atrial/fisiopatología , Vías Autónomas/fisiología , Bradicardia/fisiopatología , Perros , Estimulación Eléctrica , Femenino , Ganglios Autónomos/efectos de los fármacos , Ganglios Autónomos/fisiopatología , Bloqueadores Ganglionares/farmacología , Corazón/efectos de los fármacos , Corazón/fisiopatología , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/inervación , Atrios Cardíacos/fisiopatología , Hexametonio/farmacología , Masculino , Modelos Animales , Neuronas/fisiología
16.
eNeuro ; 9(2)2022.
Artículo en Inglés | MEDLINE | ID: mdl-35115383

RESUMEN

Axon guidance receptors such as deleted in colorectal cancer (DCC) contribute to the normal formation of neural circuits, and their mutations can be associated with neural defects. In humans, heterozygous mutations in DCC have been linked to congenital mirror movements, which are involuntary movements on one side of the body that mirror voluntary movements of the opposite side. In mice, obvious hopping phenotypes have been reported for bi-allelic Dcc mutations, while heterozygous mutants have not been closely examined. We hypothesized that a detailed characterization of Dcc heterozygous mice may reveal impaired corticospinal and spinal functions. Anterograde tracing of the Dcc+/- motor cortex revealed a normally projecting corticospinal tract, intracortical microstimulation (ICMS) evoked normal contralateral motor responses, and behavioral tests showed normal skilled forelimb coordination. Gait analyses also showed a normal locomotor pattern and rhythm in adult Dcc+/- mice during treadmill locomotion, except for a decreased occurrence of out-of-phase walk and an increased duty cycle of the stance phase at slow walking speed. Neonatal isolated Dcc+/- spinal cords had normal left-right and flexor-extensor coupling, along with normal locomotor pattern and rhythm, except for an increase in the flexor-related motoneuronal output. Although Dcc+/- mice do not exhibit any obvious bilateral impairments like those in humans, they exhibit subtle motor deficits during neonatal and adult locomotion.


Asunto(s)
Locomoción , Tractos Piramidales , Animales , Receptor DCC/genética , Heterocigoto , Locomoción/genética , Ratones , Neuronas Motoras/fisiología , Fenotipo
17.
Brain Res ; 1769: 147625, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34416255

RESUMEN

The nucleus of the solitary tract (NTS) receives viscerosensory information from the vagus nerve to regulate diverse homeostatic reflex functions. The NTS projects to a wide network of other brain regions, including the paraventricular nucleus of the hypothalamus (PVN). Here we examined the synaptic characteristics of primary afferent pathways to PVN-projecting NTS neurons in rat brainstem slices.Expression of the Transient Receptor Potential Vanilloid receptor (TRPV1+ ) distinguishes C-fiber afferents within the solitary tract (ST) from A-fibers (TRPV1-). We used resiniferatoxin (RTX), a TRPV1 agonist, to differentiate the two. The variability in the latency (jitter) of evoked excitatory postsynaptic currents (ST-EPSCs) distinguished monosynaptic from polysynaptic ST-EPSCs. Rhodamine injected into PVN was retrogradely transported to identify PVN-projecting NTS neurons within brainstem slices. Graded shocks to the ST elicited all-or-none EPSCs in rhodamine-positive NTS neurons with latencies that had either low jitter (<200 µs - monosynaptic), high jitter (>200 µs - polysynaptic inputs) or both. RTX blocked ST-evoked TRPV1 + EPSCs whether mono- or polysynaptic. Most PVN-projecting NTS neurons (17/21 neurons) had at least one input polysynaptically connected to the ST. Compared to unlabeled NTS neurons, PVN-projecting NTS neurons were more likely to receive indirect inputs and be higher order. Surprisingly, sEPSC rates for PVN-projecting neurons were double that of unlabeled NTS neurons. The ST synaptic responses for PVN-projecting NTS neurons were either all TRPV1+ or all TRPV1-, including neurons that received both direct and indirect inputs. Overall, PVN-projecting NTS neurons received direct and indirect vagal afferent information with strict segregation regarding TRPV1 expression.


Asunto(s)
Vías Aferentes/fisiología , Fibras Nerviosas Amielínicas/fisiología , Núcleo Hipotalámico Paraventricular/fisiología , Nervio Vago/fisiología , Animales , Diterpenos/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Masculino , Núcleo Hipotalámico Paraventricular/citología , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Núcleo Solitario/metabolismo , Sinapsis/efectos de los fármacos , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/metabolismo , Nervio Vago/citología
18.
Opt Express ; 18(10): 10068-77, 2010 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-20588860

RESUMEN

Intrinsic optical imaging (IOI) has emerged as a very powerful tool to assess neuronal function in small animals. Although it has been used extensively in the brain, its application to the spinal cord is rare. The inability of intrinsic optical techniques to resolve different depths and embedded gray matter hampers their capacity to distinguish larger vasculature contributions of hemodynamic signals originating from motoneuron and interneuron activation. Laminar optical tomography (LOT) is a recently-developed method that fills the gap left between IOI and diffuse optical imaging. With distinct source-detector separations, light that propagates deeper into tissues can be distinguished from light originating from the surface, providing depth sensitivity. In this work, LOT is investigated for the first time to image spinal cord activation with simultaneous IOI of the cortex in rats. Such proof of concept provides a powerful imaging modality to study spinal cord activation and disruption after injury.


Asunto(s)
Aumento de la Imagen/instrumentación , Médula Espinal/irrigación sanguínea , Médula Espinal/fisiología , Tomografía Óptica/instrumentación , Animales , Velocidad del Flujo Sanguíneo/fisiología , Diseño de Equipo , Análisis de Falla de Equipo , Femenino , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Médula Espinal/citología
19.
Front Physiol ; 11: 865, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32792982

RESUMEN

Modest recovery of somatic function after incomplete spinal cord injury (SCI) has been widely demonstrated. Recently we have shown that spontaneous recovery of baroreflex regulation of sympathetic activity also occurs in rats. Dietary restriction in the form of every other day fasting (EODF) has been shown to have beneficial effects on the recovery of motor function after SCI in rats. The goal of this study was to determine if EODF augments the improvement of baroreflex regulation of sympathetic activity after chronic left thoracic (T8) surgical spinal hemisection. To determine this, we performed baroreflex tests on ad-lib fed or EODF rats 1 week or 7 weeks after left T8 spinal hemisection. One week after T8 left hemisection baroreflex testing revealed that gain of baroreflex responsiveness, as well as the ability to increase renal sympathetic nerve activity (RSNA) at low arterial pressure, was significantly impaired in the ad-lib fed but not the EODF rats compared with sham lesioned control rats. However, baroreflex tests performed 7 weeks after T8 left hemisection revealed the inability of both ad-lib and EODF rats to decrease RSNA at elevated arterial pressures. While there is evidence to suggest that EODF has beneficial effects on the recovery of motor function in rats, EODF did not significantly improve the recovery of baroreflex regulation of sympathetic activity.

20.
Restor Neurol Neurosci ; 27(4): 297-305, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19738323

RESUMEN

PURPOSE: Nerve regeneration and functional recovery are often incomplete after peripheral nerve damage. The aim of this study was to determine if chondroitinase ABC injection at the lesion site, 1 hour of electrical stimulation and the combination of these treatments at the time of repair could be effective in promoting muscle reinnervation. METHODS: The right sciatic nerve was completely sectioned in 32 female Sprague-Dawley rats. End-to-end microsuture repair was undertaken and fibrin glue was added. Five groups were studied: 1) suture (S) + fibrin glue (F) only; 2) S + F + chondroitinase ABC; 3) S + F + electrical stimulation; 4) S + F + chondroitinase ABC + electrical stimulation; 5) uninjured nerve. RESULTS: Post recovery kinematics showed larger excursion of the hip-ankle-toe angle during walking in groups 2, 3 and 4 than in group 1 (p < 0.05). In vivo electromyographic activity and maximal muscle force were similar between groups 2, 3, 4 and 5, with higher values in all of them compared to group 1 (p < 0.05). Also, the distal stump of the sciatic nerve was excised, and cross-sectioning revealed that the number of axons were similar in all groups. CONCLUSIONS: 150 days after nerve transection, recovery was incomplete with S and F only. Chondroitinase ABC injection at the lesion site and/or 1 hour of electrical stimulation of the proximal nerve stump were beneficial in promoting nerve regeneration and functional muscle reinnervation.


Asunto(s)
Condroitina ABC Liasa/uso terapéutico , Estimulación Eléctrica , Músculo Esquelético/inervación , Músculo Esquelético/fisiopatología , Neuropatía Ciática/fisiopatología , Neuropatía Ciática/terapia , Animales , Fenómenos Biomecánicos , Condroitina ABC Liasa/farmacología , Modelos Animales de Enfermedad , Electromiografía/métodos , Femenino , Fuerza Muscular/efectos de los fármacos , Fuerza Muscular/fisiología , Músculo Esquelético/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/fisiología , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/fisiología , Neuropatía Ciática/tratamiento farmacológico , Neuropatía Ciática/patología , Caminata/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA