Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.773
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Eur J Neurosci ; 60(5): 4893-4906, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39073048

RESUMEN

We investigated the impact of the human-specific gene CHRFAM7A on the function of α7 nicotinic acetylcholine receptors (α7 nAChRs) in two different types of neurons: human-induced pluripotent stem cell (hiPSC)-derived cortical neurons, and superior cervical ganglion (SCG) neurons, taken from transgenic mice expressing CHRFAM7A. dupα7, the gene product of CHRFAM7A, which lacks a major part of the extracellular N-terminal ligand-binding domain, co-assembles with α7, the gene product of CHRNA7. We assessed the receptor function in hiPSC-derived cortical and SCG neurons with Fura-2 calcium imaging and three different α7-specific ligands: PNU282987, choline, and 4BP-TQS. Given the short-lived open state of α7 receptors, we combined the two orthosteric agonists PNU282987 and choline with the type-2 positive allosteric modulator (PAM II) PNU120596. In line with different cellular models used previously, we demonstrate that CHRFAM7A has a major impact on nicotinic α7 nAChRs by reducing calcium transients in response to all three agonists.


Asunto(s)
Células Madre Pluripotentes Inducidas , Ratones Transgénicos , Neuronas , Receptor Nicotínico de Acetilcolina alfa 7 , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/genética , Animales , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Humanos , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Ratones , Colina/farmacología , Colina/metabolismo , Ganglio Cervical Superior/citología , Ganglio Cervical Superior/metabolismo , Compuestos Bicíclicos con Puentes/farmacología , Agonistas Nicotínicos/farmacología , Benzamidas/farmacología , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Corteza Cerebral/efectos de los fármacos , Calcio/metabolismo , Isoxazoles , Compuestos de Fenilurea
2.
Clin Auton Res ; 34(1): 45-77, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38393672

RESUMEN

PURPOSE: The heart receives cervical and thoracic sympathetic contributions. Although the stellate ganglion is considered the main contributor to cardiac sympathetic innervation, the superior cervical ganglia (SCG) is used in many experimental studies. The clinical relevance of the SCG to cardiac innervation is controversial. We investigated current morphological and functional evidence as well as controversies on the contribution of the SCG to cardiac innervation. METHODS: A systematic literature review was conducted in PubMed, Embase, Web of Science, and COCHRANE Library. Included studies received a full/text review and quality appraisal. RESULTS: Seventy-six eligible studies performed between 1976 and 2023 were identified. In all species studied, morphological evidence of direct or indirect SCG contribution to cardiac innervation was found, but its contribution was limited. Morphologically, SCG sidedness may be relevant. There is indirect functional evidence that the SCG contributes to cardiac innervation as shown by its involvement in sympathetic overdrive reactions in cardiac disease states. A direct functional contribution was not found. Functional data on SCG sidedness was largely unavailable. Information about sex differences and pre- and postnatal differences was lacking. CONCLUSION: Current literature mainly supports an indirect involvement of the SCG in cardiac innervation, via other structures and plexuses or via sympathetic overdrive in response to cardiac diseases. Morphological evidence of a direct involvement was found, but its contribution seems limited. The relevance of SCG sidedness, sex, and developmental stage in health and disease remains unclear and warrants further exploration.


Asunto(s)
Corazón , Ganglio Cervical Superior , Humanos , Ganglio Cervical Superior/fisiología , Corazón/inervación , Corazón/fisiología , Animales , Sistema Nervioso Autónomo/fisiología , Cardiopatías/fisiopatología
3.
Int J Mol Sci ; 25(5)2024 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-38473838

RESUMEN

The occurrence, inhibitory modulation, and trophic effects of GABA have been identified in the peripheral sympathetic nervous system. We have demonstrated that GABA and acetylcholine (ACh) may colocalize in the same axonal varicosities or be segregated into separate ones in the rat superior cervical ganglia (SCG). Neurotransmitter segregation varies with age and the presence of neurotrophic factors. Here, we explored age-dependent changes in the occurrence and segregation of GABA and ACh in rats ranging from 2 weeks old (wo) to 12 months old or older. Using immunohistochemistry, we characterized the expression of L-glutamic acid decarboxylase of 67 kDa (GAD67) and vesicular acetylcholine transporter (VAChT) in the rat SCG at 2, 4, 8, 12 wo and 12 months old or older. Our findings revealed that GAD67 was greater at 2 wo compared with the other ages, whereas VAChT levels were greater at 4 wo than at 12 wo and 12 months old or older. The segregation of these neurotransmitters was more pronounced at 2 and 4 wo. We observed a caudo-rostral gradient of segregation degree at 8 and 12 wo. Data point out that the occurrence and segregation of GABA and ACh exhibit developmental adaptative changes throughout the lifetime of rats. We hypothesize that during the early postnatal period, the increase in GABA and GABA-ACh segregation promotes the release of GABA alone which might play a role in trophic actions.


Asunto(s)
Acetilcolina , Ganglio Cervical Superior , Ratas , Animales , Acetilcolina/metabolismo , Axones/metabolismo , Ácido gamma-Aminobutírico/metabolismo
4.
Can J Physiol Pharmacol ; 101(10): 539-547, 2023 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-37406358

RESUMEN

Ganglionic long-term potentiation (gLTP) in the rat superior cervical ganglion (SCG) is differentially modulated by neurotrophic factors (Nts): brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF). KCNQ/M channels, key regulators of neuronal excitability, and firing pattern are modulated by Nts; therefore, they might contribute to gLTP expression and to the Nts-dependent modulation of gLTP. In the SCG of rats, we characterized the presence of the KCNQ2 isoform and the effects of opposite KCNQ/M channel modulators on gLTP in control condition and under Nts modulation. Immunohistochemical and reverse transcriptase polymerase chain reaction analyses showed the expression of the KCNQ2 isoform. We found that 1 µmol/L XE991, a channel inhibitor, significantly reduced gLTP (∼50%), whereas 5 µmol/L flupirtine, a channel activator, significantly increased gLTP (1.3- to 1.7-fold). Both modulators counterbalanced the effects of the Nts on gLTP. Data suggest that KCNQ/M channels are likely involved in gLTP expression and in the modulation exerted by BDNF and NGF.


Asunto(s)
Potenciación a Largo Plazo , Ganglio Cervical Superior , Ratas , Animales , Ganglio Cervical Superior/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor de Crecimiento Nervioso/farmacología , Transducción de Señal
5.
Circ Res ; 126(6): 708-721, 2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-31928179

RESUMEN

RATIONALE: Effector memory T lymphocytes (TEM cells) exacerbate hypertension in response to repeated hypertensive stimuli. These cells reside in the bone marrow for prolonged periods and can be reactivated on reexposure to the hypertensive stimulus. OBJECTIVE: Because hypertension is associated with increased sympathetic outflow to the bone marrow, we hypothesized that sympathetic nerves regulate accumulation and reactivation of bone marrow-residing hypertension-specific TEM cells. METHODS AND RESULTS: Using unilateral superior cervical ganglionectomy in wild-type C57BL/6 mice, we showed that sympathetic nerves create a bone marrow environment that supports residence of hypertension-specific CD8+ T cells. These cells, defined by their proliferative response on coculture with dendritic cells from Ang (angiotensin) II-infused mice, were reduced in denervated compared with innervated bone of Ang II-infused mice. Adoptively transferred CD8+ T cells from Ang II-infused mice preferentially homed to innervated compared with denervated bone. In contrast, ovalbumin responsive T cells from OT-I mice did not exhibit this preferential homing. Increasing superior cervical ganglion activity by activating Gq-coupled designer receptor exclusively activated by designer drug augmented CD8+ TEM bone marrow accumulation. Adoptive transfer studies using mice lacking ß2AR (ß2 adrenergic receptors) indicate that ß2AR in the bone marrow niche, rather than T-cell ß2AR is critical for TEM cell homing. Inhibition of global sympathetic outflow using Gi-coupled DREADD (designer receptor exclusively activated by designer drug) injected into the rostral ventrolateral medulla or treatment with a ß2AR antagonist reduced hypertension-specific CD8+ TEM cells in the bone marrow and reduced the hypertensive response to a subsequent response to low dose Ang II. CONCLUSIONS: Sympathetic nerves contribute to the homing and survival of hypertension-specific TEM cells in the bone marrow after they are formed in hypertension. Inhibition of sympathetic nerve activity and ß2AR blockade reduces these cells and prevents the blood pressure elevation and renal inflammation on reexposure to hypertension stimuli.


Asunto(s)
Médula Ósea/inervación , Linfocitos T CD8-positivos/fisiología , Movimiento Celular , Hipertensión/fisiopatología , Ganglio Cervical Superior/fisiopatología , Traslado Adoptivo , Antagonistas de Receptores Adrenérgicos beta 2/farmacología , Angiotensina II/farmacología , Animales , Médula Ósea/inmunología , Linfocitos T CD8-positivos/inmunología , Desnervación , Hipertensión/inmunología , Bulbo Raquídeo/efectos de los fármacos , Bulbo Raquídeo/fisiopatología , Ratones , Ratones Endogámicos C57BL , Receptores Adrenérgicos beta 2/metabolismo , Ganglio Cervical Superior/efectos de los fármacos
6.
J Pharmacol Sci ; 148(1): 103-107, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34924113

RESUMEN

We have reported that nicotine has a neurotrophic action on peripheral adrenergic nerves in vivo, which is mediated by α7 nicotinic acetylcholine receptors (nAChRs). To clarify the possible mechanisms, the present study further investigated the effect of nicotine on neurite outgrowth in tyrosine hydroxylase (TH)-positive superior cervical ganglia (SCG) cells isolated from neonatal rats in vitro. Nicotine at low concentrations (0.01-0.3 mM) increased the number of neurite outgrowths in TH-immunopositive SCG cells, while high concentrations of nicotine (1-10 mM) gradually reduced it, and only 10 mM nicotine was markedly inhibited compared to the control. A 100 µM of nicotine-induced increase in neurite numbers depended on the exposure time and was inhibited by treatment with the nAChR antagonist hexamethonium (Hex) and α7 nAChR antagonist α-bungarotoxin (α-Bgtx). The nicotine (10 mM)-induced a significant decrease in neurite outgrowth in SCG, which was perfectly canceled by Hex to the control level but not by α-Bgtx. These results suggest that nicotine has a regulatory neurotrophic action mediated by both α7 nAChR and other subtypes in TH-positive SCG cells of rats.


Asunto(s)
Factores de Crecimiento Nervioso , Neuritas/efectos de los fármacos , Neuritas/fisiología , Proyección Neuronal/efectos de los fármacos , Nicotina/farmacología , Ganglio Cervical Superior/citología , Ganglio Cervical Superior/fisiología , Animales , Células Cultivadas , Ratas , Receptor Nicotínico de Acetilcolina alfa 7/fisiología
7.
Morphologie ; 106(352): 4-7, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33485782

RESUMEN

The jugular nerve (JN) is described as joining the superior cervical ganglion to the vagus nerve. It has been studied extensively in many different animal species; however, there is very limited literature about humans. This review delves into various descriptions of this nerve's anatomy and animal studies aimed at deciphering its function. The goal is to shed more light on this understudied structure in humans.


Asunto(s)
Nervio Glosofaríngeo , Ganglio Cervical Superior , Animales , Humanos , Nervio Vago
8.
Development ; 145(22)2018 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-30337376

RESUMEN

TWE-PRIL is a naturally occurring fusion protein of components of two TNF superfamily members: the extracellular domain of APRIL; and the intracellular and transmembrane domains of TWEAK with no known function. Here, we show that April-/- mice (which lack APRIL and TWE-PRIL) exhibited overgrowth of sympathetic fibres in vivo, and sympathetic neurons cultured from these mice had significantly longer axons than neurons cultured from wild-type littermates. Enhanced axon growth from sympathetic neurons cultured from April-/- mice was prevented by expressing full-length TWE-PRIL in these neurons but not by treating them with soluble APRIL. Soluble APRIL, however, enhanced axon growth from the sympathetic neurons of wild-type mice. siRNA knockdown of TWE-PRIL but not siRNA knockdown of APRIL alone also enhanced axon growth from wild-type sympathetic neurons. Our work reveals the first and physiologically relevant role for TWE-PRIL and suggests that it mediates reverse signalling.


Asunto(s)
Axones/metabolismo , Transducción de Señal , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Animales , Antígeno de Maduración de Linfocitos B/genética , Antígeno de Maduración de Linfocitos B/metabolismo , Células Cultivadas , Citocina TWEAK/genética , Citocina TWEAK/metabolismo , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Técnicas de Silenciamiento del Gen , Ratones , Modelos Biológicos , Factor de Crecimiento Nervioso/farmacología , Fenotipo , ARN Interferente Pequeño/metabolismo , Solubilidad , Ganglio Cervical Superior/metabolismo , Sistema Nervioso Simpático/crecimiento & desarrollo , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
9.
Neuroradiology ; 62(4): 519-524, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31996966

RESUMEN

PURPOSE: This study aimed to assess the magnetic resonance (MRI) features of the superior cervical ganglion (SCG) and to track changes to it induced using radiotherapy across a long-term follow-up. METHODS: In total, 75 patients who underwent radiotherapy for head and neck malignancies and who were studied with MRI were recruited from two centers. MRI was performed before and after radiotherapy, with a median long-term follow-up of 4.5 years. Baseline SCG features were assessed. Changes in axial cross-sectional area, T2-normalized signal, and apparent diffusion coefficient (ADC) (the latter available in about half of the patients) were analyzed. Repeated measures analysis of variance with Bonferroni's correction was used to analyze changes in the aforementioned parameters (significance level 0.05). RESULTS: Out of a potential 149 SCGs, 136 were visible at baseline MRI. A variable spatial relationship with the internal carotid artery was found. SCGs showed the "black dot" sign in almost all of the patients. ADC was higher in SCGs than in regional lymph nodes. Cross-sectional area, normalized T2, and ADC increased in the period up to 1 year after radiotherapy and then remained stable in subsequent longer-term follow-up. CONCLUSION: The SCG has unusual features that allow differentiation from the regional lymph nodes. Changes in morphology and signal after radiotherapy must be taken into account by radiologists to avoid misdiagnosis as recurrent nodal disease. Changes induced using radiotherapy are stable in long-term follow-up and are thus likely attributed to other factors (such as Schwann cell hypertrophy/proliferation) rather than edema.


Asunto(s)
Neoplasias de Cabeza y Cuello/diagnóstico por imagen , Neoplasias de Cabeza y Cuello/radioterapia , Imagen por Resonancia Magnética/métodos , Ganglio Cervical Superior/diagnóstico por imagen , Ganglio Cervical Superior/efectos de la radiación , Anciano , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
10.
Biol Res ; 53(1): 31, 2020 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-32650839

RESUMEN

BACKGROUND: In modern societies, sleep deprivation is a serious health problem. This problem could be induced by a variety of reasons, including lifestyle habits or neurological disorders. Chronic sleep deprivation (CSD) could have complex biological consequences, such as changes in neural autonomic control, increased oxidative stress, and inflammatory responses. The superior cervical ganglion (SCG) is an important sympathetic component of the autonomic nervous system. CSD can lead to a wide range of neurological consequences in SCG, which mainly supply innervations to circadian system and other structures. As the active component of Curcuma longa, curcumin possesses many therapeutic properties; including neuroprotective. This study aimed to evaluate the effect of CSD on the SCG histomorphometrical changes and the protective effect of curcumin in preventing these changes. METHODS: Thirty-six male rats were randomly assigned to the control, curcumin, CSD, CSD + curcumin, grid floor control, and grid floor + curcumin groups. The CSD was induced by a modified multiple platform apparatus for 21 days and animals were sacrificed at the end of CSD or treatment, and their SCGs removed for stereological and TUNEL evaluations and also spatial arrangement of neurons in this structure. RESULTS: Concerning stereological findings, CSD significantly reduced the volume of SCG and its total number of neurons and satellite glial cells in comparison with the control animals (P < 0.05). Treatment of CSD with curcumin prevented these decreases. Furthermore, TUNEL evaluation showed significant apoptosis in the SCG cells in the CSD group, and treatment with curcumin significantly decreased this apoptosis (P < 0.01). This decrease in apoptosis was observed in all control groups that received curcumin. CSD also changed the spatial arrangement of ganglionic neurons into a random pattern, whereas treatment with curcumin preserved its regular pattern. CONCLUSIONS: CSD could potentially induce neuronal loss and structural changes including random spatial distribution in the SCG neurons. Deleterious effects of sleep deprivation could be prevented by the oral administration of curcumin. Furthermore, the consumption of curcumin in a healthy person might lead to a reduction of cell death.


Asunto(s)
Antiinflamatorios no Esteroideos , Curcumina , Privación de Sueño , Ganglio Cervical Superior , Animales , Antiinflamatorios no Esteroideos/farmacología , Curcumina/farmacología , Masculino , Ratas , Ratas Sprague-Dawley , Privación de Sueño/tratamiento farmacológico , Privación de Sueño/patología , Ganglio Cervical Superior/efectos de los fármacos
11.
Int J Mol Sci ; 21(16)2020 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-32806753

RESUMEN

The ionic mechanisms controlling the resting membrane potential (RMP) in superior cervical ganglion (SCG) neurons have been widely studied and the M-current (IM, KCNQ) is one of the key players. Recently, with the discovery of the presence of functional TREK-2 (TWIK-related K+ channel 2) channels in SCG neurons, another potential main contributor for setting the value of the resting membrane potential has appeared. In the present work, we quantified the contribution of TREK-2 channels to the resting membrane potential at physiological temperature and studied its role in excitability using patch-clamp techniques. In the process we have discovered that TREK-2 channels are sensitive to the classic M-current blockers linopirdine and XE991 (IC50 = 0.310 ± 0.06 µM and 0.044 ± 0.013 µM, respectively). An increase from room temperature (23 °C) to physiological temperature (37 °C) enhanced both IM and TREK-2 currents. Likewise, inhibition of IM by tetraethylammonium (TEA) and TREK-2 current by XE991 depolarized the RMP at room and physiological temperatures. Temperature rise also enhanced adaptation in SCG neurons which was reduced due to TREK-2 and IM inhibition by XE991 application. In summary, TREK-2 and M currents contribute to the resting membrane potential and excitability at room and physiological temperature in the primary culture of mouse SCG neurons.


Asunto(s)
Canales de Potasio KCNQ/metabolismo , Potenciales de la Membrana , Neuronas/fisiología , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Sistema Nervioso Simpático/fisiología , Temperatura , Adaptación Fisiológica/efectos de los fármacos , Animales , Antracenos/farmacología , Células HEK293 , Humanos , Indoles/farmacología , Activación del Canal Iónico/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Ratones , Neuronas/efectos de los fármacos , Piridinas/farmacología , Riluzol/farmacología , Ganglio Cervical Superior/efectos de los fármacos , Ganglio Cervical Superior/fisiología , Tetraetilamonio/farmacología , Tetrahidronaftalenos/farmacología , Tetrazoles/farmacología
12.
Int J Mol Sci ; 21(2)2020 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-31936257

RESUMEN

Bradykinin (BK), a hormone inducing pain and inflammation, is known to inhibit potassium M-currents (IM) and to increase the excitability of the superior cervical ganglion (SCG) neurons by activating the Ca2+-calmodulin pathway. M-current is also reduced by muscarinic agonists through the depletion of membrane phosphatidylinositol 4,5-biphosphate (PIP2). Similarly, the activation of muscarinic receptors inhibits the current through two-pore domain potassium channels (K2P) of the "Tandem of pore-domains in a Weakly Inward rectifying K+ channel (TWIK)-related channels" (TREK) subfamily by reducing PIP2 in mouse SCG neurons (mSCG). The aim of this work was to test and characterize the modulation of TREK channels by bradykinin. We used the perforated-patch technique to investigate riluzole (RIL) activated currents in voltage- and current-clamp experiments. RIL is a drug used in the palliative treatment of amyotrophic lateral sclerosis and, in addition to blocking voltage-dependent sodium channels, it also selectively activates the K2P channels of the TREK subfamily. A cell-attached patch-clamp was also used to investigate TREK-2 single channel currents. We report here that BK reduces spike frequency adaptation (SFA), inhibits the riluzole-activated current (IRIL), which flows mainly through TREK-2 channels, by about 45%, and reduces the open probability of identified single TREK-2 channels in cultured mSCG cells. The effect of BK on IRIL was precluded by the bradykinin receptor (B2R) antagonist HOE-140 (d-Arg-[Hyp3, Thi5, d-Tic7, Oic8]BK) but also by diC8PIP2 which prevents PIP2 depletion when phospholipase C (PLC) is activated. On the contrary, antagonizing inositol triphosphate receptors (IP3R) using 2-aminoethoxydiphenylborane (2-APB) or inhibiting protein kinase C (PKC) with bisindolylmaleimide did not affect the inhibition of IRIL by BK. In conclusion, bradykinin inhibits TREK-2 channels through the activation of B2Rs resulting in PIP2 depletion, much like we have demonstrated for muscarinic agonists. This mechanism implies that TREK channels must be relevant for the capture of information about pain and visceral inflammation.


Asunto(s)
Bradiquinina/metabolismo , Neuronas/efectos de los fármacos , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canales de Potasio de Dominio Poro en Tándem/genética , Sistema Nervioso Simpático/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Bradiquinina/administración & dosificación , Bradiquinina/análogos & derivados , Bradiquinina/genética , Bradiquinina/farmacología , Células Cultivadas , Humanos , Ratones , Agonistas Muscarínicos/farmacología , Neuronas/patología , Técnicas de Placa-Clamp , Fosfatidilinositol 4,5-Difosfato/genética , Potasio/metabolismo , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Receptores Muscarínicos/genética , Riluzol/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Ganglio Cervical Superior/efectos de los fármacos , Sistema Nervioso Simpático/metabolismo , Fosfolipasas de Tipo C
13.
J Neurophysiol ; 122(6): 2591-2600, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31642403

RESUMEN

NaV1.8 channels play a crucial role in regulating the action potential in nociceptive neurons. A single nucleotide polymorphism in the human NaV1.8 gene SCN10A, A1073V (rs6795970, G>A), has been linked to the diminution of mechanical pain sensation as well as cardiac conduction abnormalities. Furthermore, studies have suggested that this polymorphism may result in a "loss-of-function" phenotype. In the present study, we performed genomic analysis of A1073V polymorphism presence in a cohort of patients undergoing sigmoid colectomy who provided information regarding perioperative pain and analgesic use. Homozygous carriers reported significantly reduced severity in postoperative abdominal pain compared with heterozygous and wild-type carriers. Homozygotes also trended toward using less analgesic/opiates during the postoperative period. We also heterologously expressed the wild-type and A1073V variant in rat superior cervical ganglion neurons. Electrophysiological testing demonstrated that the mutant NaV1.8 channels activated at more depolarized potentials compared with wild-type channels. Our study revealed that postoperative abdominal pain is diminished in homozygous carriers of A1073V and that this is likely due to reduced transmission of action potentials in nociceptive neurons. Our findings reinforce the importance of NaV1.8 and the A1073V polymorphism to pain perception. This information could be used to develop new predictive tools to optimize patient pain experience and analgesic use in the perioperative setting.NEW & NOTEWORTHY We present evidence that in a cohort of patients undergoing sigmoid colectomy, those homozygous for the NaV1.8 polymorphism (rs6795970) reported significantly lower abdominal pain scores than individuals with the homozygous wild-type or heterozygous genotype. In vitro electrophysiological recordings also suggest that the mutant NaV1.8 channel activates at more depolarizing potentials than the wild-type Na+ channel, characteristic of hypoactivity. This is the first report linking the rs6795970 mutation with postoperative abdominal pain in humans.


Asunto(s)
Dolor Abdominal/genética , Colectomía , Fenómenos Electrofisiológicos/fisiología , Ganglios Espinales/fisiología , Canal de Sodio Activado por Voltaje NAV1.8/fisiología , Nocicepción/fisiología , Dolor Postoperatorio/genética , Ganglio Cervical Superior/metabolismo , Sistema Nervioso Simpático/fisiología , Anciano , Animales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Canal de Sodio Activado por Voltaje NAV1.8/genética , Neuronas/fisiología , Polimorfismo Genético , Ratas , Estudios Retrospectivos
14.
J Neurochem ; 150(6): 666-677, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31306490

RESUMEN

In the sympatho-adrenal system, angiotensin II (Ang II) acts as a key neuromodulatory component. At sympathetic nerve terminals, Ang II influences sympathetic transmission by enhancing norepinephrine (NE) synthesis, facilitating NE release and inhibiting NE uptake. Previously, it was demonstrated that tyrosine hydroxylase (TH) mRNA is trafficked to the distal axons of primary superior cervical ganglia (SCG) neurons, directed by a cis-acting regulatory element (i.e. zipcode) located in the 3'UTR of the transcript. Results of metabolic labeling studies established that the mRNA is locally translated. It was further shown that the axonal trafficking of the mRNA encoding the enzyme plays an important role in mediating dopamine (DA) and NE synthesis and may facilitate the maintenance of axonal catecholamine levels. In the present study, the hypothesis was tested that Ang II induces NE synthesis in rat primary SCG neurons via the modulation of the trafficking of the mRNAs encoding the catecholamine synthesizing enzymes TH and dopamine ß-hydroxylase (DBH). Treatment of SCG neurons with the Ang II receptor type 1 (AT1R) agonist, L-162,313, increases the axonal levels of TH and DBH mRNA and protein and results in elevated NE levels. Conversely, treatment of rat SCG neurons with the AT1R antagonist, Eprosartan, abolished the L-162,313-mediated increase in axonal levels of TH and DBH mRNA and protein. In a first attempt to identify the proteins involved in the Ang II-mediated axonal transport of TH mRNA, we used a biotinylated 50-nucleotide TH RNA zipcode as bait in the affinity purification of TH zipcode-associated proteins. Mass spectrometric analysis of the TH zipcode ribonucleoprotein (RNP) complex immune-purified from SCG neurons led to the identification of 163 somal and 127 axonal proteins functionally involved in binding nucleic acids, the translational machinery or acting as subunits of cytoskeletal and motor proteins. Surprisingly, immune-purification of the TH axonal trafficking complex, results in the acquisition of DBH mRNA, suggesting that these mRNAs maybe transported to the axon together, possibly in the same RNP complex. Taken together, our results point to a novel mechanism by which Ang II participates in the regulation of axonal synthesis of NE by modulating the local trafficking and expression of TH and DBH, two key enzymes involved in the catecholamine biosynthetic pathway.


Asunto(s)
Angiotensina II/metabolismo , Axones/metabolismo , Dopamina beta-Hidroxilasa/metabolismo , Norepinefrina/biosíntesis , Tirosina 3-Monooxigenasa/metabolismo , Fibras Adrenérgicas/metabolismo , Animales , Transporte Axonal/fisiología , Células Cultivadas , Neuronas/metabolismo , Transporte de Proteínas/fisiología , ARN Mensajero , Ratas , Ratas Sprague-Dawley , Ganglio Cervical Superior/metabolismo
15.
Development ; 143(9): 1560-70, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27143756

RESUMEN

During development of the peripheral nervous system, excess neurons are generated, most of which will be lost by programmed cell death due to a limited supply of neurotrophic factors from their targets. Other environmental factors, such as 'competition factors' produced by neurons themselves, and axon guidance molecules have also been implicated in developmental cell death. Semaphorin 3A (Sema3A), in addition to its function as a chemorepulsive guidance cue, can also induce death of sensory neurons in vitro The extent to which Sema3A regulates developmental cell death in vivo, however, is debated. We show that in compartmentalized cultures of rat sympathetic neurons, a Sema3A-initiated apoptosis signal is retrogradely transported from axon terminals to cell bodies to induce cell death. Sema3A-mediated apoptosis utilizes the extrinsic pathway and requires both neuropilin 1 and plexin A3. Sema3A is not retrogradely transported in older, survival factor-independent sympathetic neurons, and is much less effective at inducing apoptosis in these neurons. Importantly, deletion of either neuropilin 1 or plexin A3 significantly reduces developmental cell death in the superior cervical ganglia. Taken together, a Sema3A-initiated apoptotic signaling complex regulates the apoptosis of sympathetic neurons during the period of naturally occurring cell death.


Asunto(s)
Apoptosis/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neuropilina-1/metabolismo , Receptores de Superficie Celular/metabolismo , Semaforina-3A/metabolismo , Ganglio Cervical Superior/embriología , Sistema Nervioso Simpático/embriología , Animales , Axones/metabolismo , Caspasa 3/metabolismo , Células Cultivadas , Ratones , Ratones Noqueados , Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/genética , Neuropilina-1/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/genética , Transducción de Señal , Ganglio Cervical Superior/citología , Ganglio Cervical Superior/fisiología
16.
Artículo en Inglés | MEDLINE | ID: mdl-30604271

RESUMEN

In chicks, axial length and choroidal thickness undergo circadian oscillations. The choroid is innervated by both branches of the autonomic nervous system, but their contribution(s) to these rhythms is unknown. We used two combination lesions to test this. For parasympathectomy, nerve VII was sectioned presynaptic to the pterygopalatine ganglia, and the ciliary post-ganglionics were cut (double lesion; n = 8). Triple lesions excised the sympathetic superior cervical ganglion as well (n = 8). Sham surgery was done in controls (n = 7). 8-14 days later, axial dimensions were measured with ultrasonography at 4-h intervals over 24 h. Rhythm parameters were assessed using a "best fit" function, and growth rates measured. Both types of lesions resulted in ultradian (> 1 cycle/24 h) rhythms in choroidal thickness and axial length, and increased vitreous chamber growth (Exp-fellow: double: 69 µm; triple: 104 µm; p < 0.05). For double lesions, the frequency was 1.5 cycles/day for both rhythms; for triples the choroidal rhythm was 1.5 cycles/day, and the axial was 3 cycles/day. For double lesions, the amplitudes of both rhythms were larger than those of sham surgery controls (axial: 107 vs 54 µm; choroid: 124 vs 29 µm, p < 0.05). These findings provide evidence for the involvement of abnormal ocular rhythms in the growth stimulation underlying myopia development.


Asunto(s)
Desnervación Autonómica , Longitud Axial del Ojo/inervación , Pollos/fisiología , Coroides/inervación , Ganglios Parasimpáticos/cirugía , Miopía/fisiopatología , Ganglio Cervical Superior/cirugía , Ritmo Ultradiano , Animales , Animales Recién Nacidos , Factores de Tiempo , Visión Ocular
17.
Cephalalgia ; 39(13): 1700-1709, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31370669

RESUMEN

PURPOSE: The pineal gland plays an important role in biological rhythms, circadian and circannual variations, which are key aspects in several headache disorders. OVERVIEW: Melatonin, the main pineal secreting hormone, has been extensively studied in primary and secondary headache disorders. Altered melatonin secretion occurs in many headache syndromes. Experimental data show pineal gland and melatonin both interfere in headache animal models, decreasing trigeminal activation. Melatonin has been shown to regulate CGRP and control its release. DISCUSSION: Melatonin has been used successfully as a treatment for migraine, cluster headaches and other headaches. There is a rationale for including the pineal gland as a relevant brain structure in the mechanisms of headache pathophysiology, and melatonin as a treatment option in primary headache.


Asunto(s)
Cefalea/fisiopatología , Glándula Pineal/fisiopatología , Adulto , Animales , Péptido Relacionado con Gen de Calcitonina/fisiología , Estudios de Casos y Controles , Niño , Ritmo Circadiano/fisiología , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Método Doble Ciego , Cefalea/diagnóstico por imagen , Cefalea/tratamiento farmacológico , Cefalea/patología , Humanos , Melatonina/fisiología , Melatonina/uso terapéutico , Oxidación-Reducción , Núcleo Hipotalámico Paraventricular/fisiopatología , Glándula Pineal/metabolismo , Glándula Pineal/patología , Receptores de Melatonina/agonistas , Receptores de Melatonina/fisiología , Serotonina/metabolismo , Ganglio Cervical Superior/fisiopatología
18.
Proc Natl Acad Sci U S A ; 113(23): E3290-9, 2016 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-27217553

RESUMEN

Myo-inositol is an important cellular osmolyte in autoregulation of cell volume and fluid balance, particularly for mammalian brain and kidney cells. We find it also regulates excitability. Myo-inositol is the precursor of phosphoinositides, key signaling lipids including phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2]. However, whether myo-inositol accumulation during osmoregulation affects signaling and excitability has not been fully explored. We found that overexpression of the Na(+)/myo-inositol cotransporter (SMIT1) and myo-inositol supplementation enlarged intracellular PI(4,5)P2 pools, modulated several PI(4,5)P2-dependent ion channels including KCNQ2/3 channels, and attenuated the action potential firing of superior cervical ganglion neurons. Further experiments using the rapamycin-recruitable phosphatase Sac1 to hydrolyze PI(4)P and the P4M probe to visualize PI(4)P suggested that PI(4)P levels increased after myo-inositol supplementation with SMIT1 expression. Elevated relative levels of PIP and PIP2 were directly confirmed using mass spectrometry. Inositol trisphosphate production and release of calcium from intracellular stores also were augmented after myo-inositol supplementation. Finally, we found that treatment with a hypertonic solution mimicked the effect we observed with SMIT1 overexpression, whereas silencing tonicity-responsive enhancer binding protein prevented these effects. These results show that ion channel function and cellular excitability are under regulation by several "physiological" manipulations that alter the PI(4,5)P2 setpoint. We demonstrate a previously unrecognized linkage between extracellular osmotic changes and the electrical properties of excitable cells.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Neuronas/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Simportadores/metabolismo , Potenciales de Acción , Señalización del Calcio , Línea Celular , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Células HEK293 , Proteínas de Choque Térmico/genética , Humanos , Inositol/metabolismo , Canal de Potasio KCNQ2/genética , Canal de Potasio KCNQ2/metabolismo , Canal de Potasio KCNQ3/genética , Canal de Potasio KCNQ3/metabolismo , Osmorregulación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ganglio Cervical Superior/citología , Ganglio Cervical Superior/metabolismo , Simportadores/genética , Canales Catiónicos TRPM/metabolismo , Factores de Transcripción/metabolismo
19.
Int J Mol Sci ; 20(18)2019 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-31500374

RESUMEN

Alcohol causes diverse acute and chronic symptoms that often lead to critical health problems. Exposure to ethanol alters the activities of sympathetic neurons that control the muscles, eyes, and blood vessels in the brain. Although recent studies have revealed the cellular targets of ethanol, such as ion channels, the molecular mechanism by which alcohol modulates the excitability of sympathetic neurons has not been determined. Here, we demonstrated that ethanol increased the discharge of membrane potentials in sympathetic neurons by inhibiting the M-type or Kv7 channel consisting of the Kv7.2/7.3 subunits, which were involved in determining the membrane potential and excitability of neurons. Three types of sympathetic neurons, classified by their threshold of activation and firing patterns, displayed distinct sensitivities to ethanol, which were negatively correlated with the size of the Kv7 current that differs depending on the type of neuron. Using a heterologous expression system, we further revealed that the inhibitory effects of ethanol on Kv7.2/7.3 currents were facilitated or diminished by adjusting the amount of plasma membrane phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2). These results suggested that ethanol and PI(4,5)P2 modulated gating of the Kv7 channel in superior cervical ganglion neurons in an antagonistic manner, leading to regulation of the membrane potential and neuronal excitability, as well as the physiological functions mediated by sympathetic neurons.


Asunto(s)
Potenciales de Acción , Etanol/metabolismo , Canales de Potasio KCNQ/metabolismo , Neuronas/fisiología , Fosfatidilinositol 4,5-Difosfato/metabolismo , Ganglio Cervical Superior/citología , Biomarcadores , Membrana Celular/metabolismo , Células Cultivadas , Etanol/farmacología , Expresión Génica , Canales de Potasio KCNQ/antagonistas & inhibidores , Canales de Potasio KCNQ/genética
20.
Acta Vet Hung ; 67(1): 98-105, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30922095

RESUMEN

Neuropeptide Y (NPY) is a neuronal active substance taking part in the regulation of gastrointestinal (GI) tract activity. This study used retrograde neuronal tracing and immunofluorescence methods to analyse NPY-positive neurons located in superior cervical ganglion and supplying the cervical oesophagus in the pig. The presence of NPY was observed in 30% of all neurons supplying the part of oesophagus studied. Probably the number of Fast Blue (FB) positive cells depends on the area of the wall injected with FB and the fragment of oesophagus studied. Therefore, the obtained results indicate that the described peptide is an important factor in the extrinsic innervation of this part of the GI tract.


Asunto(s)
Esófago/inervación , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Ganglio Cervical Superior/citología , Porcinos , Animales , Femenino , Inmunohistoquímica/métodos , Inmunohistoquímica/veterinaria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA