Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 14(1): 7473, 2024 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-38553555

RESUMEN

Orexin signaling plays a facilitatory role in respiration. Abnormalities in orexin levels correlate with disordered breathing patterns and impaired central respiratory chemoreception. Nucleus tractus solitarii (NTS) neurons expressing the transcription factor Phox2b contribute to the chemoreceptive regulation of respiration. However, the extent to which orexinergic signaling modulates respiratory activity in these Phox2b-expressing NTS neurons remains unclear. In the present study, the injection of orexin A into the NTS significantly increased the firing rate of the phrenic nerve. Further analysis using fluorescence in situ hybridization and immunohistochemistry revealed that orexin 1 receptors (OX1Rs) were primarily located in the ventrolateral subdivision of the NTS and expressed in 25% of Phox2b-expressing neurons. Additionally, electrophysiological recordings showed that exposure to orexin A increased the spontaneous firing rate of Phox2b-expressing neurons. Immunostaining experiments with cFos revealed that the OX1R-residing Phox2b-expressing neurons were activated by an 8% CO2 stimulus. Crucially, OX1R knockdown in these NTS neurons notably blunted the ventilatory response to 8% CO2, alongside an increase in sigh-related apneas. In conclusion, orexinergic signaling in the NTS facilitates breathing through the activation of OX1Rs, which induces the depolarization of Phox2b-expressing neurons. OX1Rs are essential for the involvement of Phox2b-expressing NTS neurons in the hypercapnic ventilatory response.


Asunto(s)
Dióxido de Carbono , Núcleo Solitario , Núcleo Solitario/metabolismo , Orexinas/metabolismo , Hibridación Fluorescente in Situ , Respiración
2.
Trauma Violence Abuse ; : 15248380241235639, 2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38516894

RESUMEN

Although numerous factors have been found to influence postpartum depression (PPD), no previous meta-analysis have systematically explored whether it is affected by adverse childhood experiences (ACEs). This study aimed to explore the influence of ACEs and their subtypes on PPD. A systematic literature search was conducted using Web of Science, PubMed, Embase, Wan Fang, China Science and Technology Journal Database, Chinese Biomedical Database, and China National Knowledge Infrastructure, and literature was screened according to inclusion and exclusion criteria. Methodological quality assessment and data extraction were performed on the included studies. A random-effects model was used to pool the effects. In total, 24 studies were included, and 73 independent effects were extracted from them. The meta-analysis revealed that ACE was a risk factor for PPD (odds ratio [OR] = 2.31, 95% confidence interval [CI] [2.04, 2.63]). The subgroup analysis results showed that emotional abuse was the ACE subtype most strongly related to the occurrence of PPD (OR = 2.95, 95% CI [2.08, 4.20]), followed by emotional neglect (OR = 2.87, 95% CI [1.89, 4.36]) and sexual abuse (OR = 2.81, 95% CI [1.93, 4.09]). In addition, family member incarceration (OR = 2.62, 95% CI [1.51, 4.54]), physical abuse (OR = 2.31, 95% CI [1.67, 3.19]), and physical neglect (OR = 2.15, 95% CI [1.36, 3.39]) also have strong effects on PPD. ACE is a risk factor for PPD. Early screening of ACE plays an important role in the prevention and intervention of PPD.

3.
Neurosci Bull ; 38(2): 149-165, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34212297

RESUMEN

Leptin, an adipocyte-derived peptide hormone, has been shown to facilitate breathing. However, the central sites and circuit mechanisms underlying the respiratory effects of leptin remain incompletely understood. The present study aimed to address whether neurons expressing leptin receptor b (LepRb) in the nucleus tractus solitarii (NTS) contribute to respiratory control. Both chemogenetic and optogenetic stimulation of LepRb-expressing NTS (NTSLepRb) neurons notably activated breathing. Moreover, stimulation of NTSLepRb neurons projecting to the lateral parabrachial nucleus (LPBN) not only remarkably increased basal ventilation to a level similar to that of the stimulation of all NTSLepRb neurons, but also activated LPBN neurons projecting to the preBötzinger complex (preBötC). By contrast, ablation of NTSLepRb neurons projecting to the LPBN notably eliminated the enhanced respiratory effect induced by NTSLepRb neuron stimulation. In brainstem slices, bath application of leptin rapidly depolarized the membrane potential, increased the spontaneous firing rate, and accelerated the Ca2+ transients in most NTSLepRb neurons. Therefore, leptin potentiates breathing in the NTS most likely via an NTS-LPBN-preBötC circuit.


Asunto(s)
Leptina , Núcleo Solitario , Leptina/metabolismo , Leptina/farmacología , Potenciales de la Membrana , Neuronas/metabolismo , Núcleo Solitario/metabolismo
4.
Neurosci Bull ; 37(1): 31-44, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32468398

RESUMEN

The locus coeruleus (LC) has been implicated in the control of breathing. Congenital central hypoventilation syndrome results from mutation of the paired-like homeobox 2b (Phox2b) gene that is expressed in LC neurons. The present study was designed to address whether stimulation of Phox2b-expressing LC (Phox2bLC) neurons affects breathing and to reveal the putative circuit mechanism. A Cre-dependent viral vector encoding a Gq-coupled human M3 muscarinic receptor (hM3Dq) was delivered into the LC of Phox2b-Cre mice. The hM3Dq-transduced neurons were pharmacologically activated while respiratory function was measured by plethysmography. We demonstrated that selective stimulation of Phox2bLC neurons significantly increased basal ventilation in conscious mice. Genetic ablation of these neurons markedly impaired hypercapnic ventilatory responses. Moreover, stimulation of Phox2bLC neurons enhanced the activity of preBötzinger complex neurons. Finally, axons of Phox2bLC neurons projected to the preBötzinger complex. Collectively, Phox2bLC neurons contribute to the control of breathing most likely via an LC-preBötzinger complex circuit.


Asunto(s)
Proteínas de Homeodominio , Locus Coeruleus , Animales , Proteínas de Unión al ADN , Factores de Intercambio de Guanina Nucleótido , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Locus Coeruleus/metabolismo , Ratones , Neuronas/metabolismo , Respiración , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
J Physiol ; 599(4): 1115-1130, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33347681

RESUMEN

KEY POINTS: This study demonstrates that both CO2 -induced respiratory and cardiovascular responses are augmented in spontaneously hypertensive rats (SHRs). Genetic ablation of the retrotrapezoid nucleus (RTN) neurons depresses enhanced hypercapnic ventilatory response and eliminates CO2 -stimulated increase in arterial pressure and heart rate in SHRs. SHRs have a high protein level of pH-sensitive channels in the RTN, including the TASK-2 channel, Kv12.1 channel and acid-sensing ion channel 3. The inhibition of putative TASK-2 channel activity by clofilium diminishes amplified hypercapnic ventilatory and cardiovascular responses, and reduces the number of CO2 -activated RTN neurons in SHRs. These results indicate that RTN neurons contribute to enhanced CO2 -stimulated respiratory and cardiovascular responses in SHRs. ABSTRACT: The respiratory regulation of cardiovascular activity is essential for maintaining an efficient ventilation and perfusion ratio. Activation of central respiratory chemoreceptors not only elicits a ventilatory response but also regulates sympathetic nerve activity and arterial blood pressure (ABP). The retrotrapezoid nucleus (RTN) is the most completely characterized cluster of central respiratory chemoreceptors. We hypothesize that RTN neurons contribute to augmented CO2 -stimulated respiratory and cardiovascular responses in adult spontaneously hypertensive rats (SHRs). Our findings indicate that SHRs exhibit more enhanced hypercapnic cardiorespiratory responses than age-matched normotensive Wistar-Kyoto rats. Genetic ablation of RTN neurons notably depresses an enhanced hypercapnic ventilatory response (HCVR) and eliminates a CO2 -stimulated greater increase in ABP and heart rate in SHRs. In addition, SHRs have a higher protein level of pH-sensitive channels in the RTN, including TASK-2 channels, Kv12.1 channels and acid-sensing ion channel 3. Administration of clofilium (i.p.), an unselective inhibitor of TASK-2 channels, not only significantly reduces the enhanced HCVR but also inhibits CO2 -amplified increases in ABP and heart rate in SHRs. Moreover, clofilium significantly decreases the number of CO2 -activated RTN neurons in SHRs. Taken together, we suggest that RTN neurons play an important role in enhanced hypercapnic ventilatory and cardiovascular responses in SHRs and the putative mechanism involved is associated with TASK-2 channel activity in the RTN.


Asunto(s)
Dióxido de Carbono , Células Quimiorreceptoras , Animales , Neuronas , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY
6.
J Neurosci ; 39(15): 2837-2846, 2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30626698

RESUMEN

The nucleus tractus solitarii (NTS) is implicated in the control of breathing, but the neuronal phenotype and circuit mechanism involved in such a physiological function remain incompletely understood. This study focused on the respiratory role of paired-like homeobox 2b gene (Phox2b)-expressing NTS neurons and sought to determine whether selective stimulation of this set of neurons activates breathing in male mice. A Cre-dependent vector encoding a Gq-coupled human M3 muscarinic receptor (hM3Dq) was microinjected into the NTS of Phox2b-Cre transgenic mice. The hM3Dq-transduced neurons were pharmacologically activated in conscious mice while respiratory effects were measured by plethysmography. We demonstrate that chemogenetic stimulation of Phox2b-expressing NTS neurons significantly increased baseline minute volume via an increase in respiratory frequency rather than tidal volume. Chemogenetic stimulation also synergized with moderate CO2 stimulation to enhance pulmonary ventilatory response. Selective ablation of Phox2b-expressing NTS neurons notably attenuated a hypercapnic ventilatory response. Moreover, histological evidence revealed that stimulation of Phox2b-expressing NTS neurons increased neuronal activity of the preBötzinger complex. Finally, we presented the neuroanatomical evidence of direct projection of Phox2b-expressing NTS neurons to putative respiratory central pattern generator. Overall, these findings suggest that selective activation of Phox2b-expressing NTS neurons potentiates baseline pulmonary ventilation via an excitatory drive to respiratory central pattern generator and this group of neurons is also required for the hypercapnic ventilatory response.SIGNIFICANCE STATEMENT The nucleus tractus solitarii (NTS) has been implicated in the control of breathing. The paired-like homeobox 2b gene (Phox2b) is the disease-defining gene for congenital central hypoventilation syndrome and is restrictively present in brainstem nucleus, including the NTS. Using a chemogenetic approach, we demonstrate herein that selective stimulation of Phox2b-expressing NTS neurons vigorously potentiates baseline pulmonary ventilation via an excitatory drive to respiratory central pattern generator in rodents. Genetic ablation of these neurons attenuates the hypercapnic ventilatory response. We also suggest that a fraction of Phox2b-expressing neurons exhibit CO2 sensitivity and presumably function as central respiratory chemoreceptors. The methodology is expected to provide a future applicability to the patients with sleep-related hypoventilation or apnea.


Asunto(s)
Proteínas de Homeodominio/fisiología , Neuronas/metabolismo , Respiración , Núcleo Solitario/metabolismo , Factores de Transcripción/fisiología , Animales , Dióxido de Carbono/farmacología , Generadores de Patrones Centrales , Fenómenos Electrofisiológicos/genética , Fenómenos Electrofisiológicos/fisiología , Proteínas de Homeodominio/genética , Hipercapnia/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microinyecciones , Pruebas de Función Respiratoria , Mecánica Respiratoria , Núcleo Solitario/citología , Factores de Transcripción/genética
7.
Can J Physiol Pharmacol ; 96(8): 807-814, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29400080

RESUMEN

Many studies have demonstrated that chronic intermittent hypobaric hypoxia (CIHH) can reduce blood pressure in spontaneously hypertensive rats and renovascular hypertensive (RVH) rats in which endothelial dysfunction is determined as a critical factor. However, whether CIHH can regulate vasodilation of the aorta in RVH rats remains unknown. The purpose of this study was to investigate the effect of CIHH on impaired relaxation of the aorta in the 2-kidney, 1-clip (2K1C) RVH rat model. The results showed CIHH improved the impaired endothelium-dependent relaxation in the 2K1C rat aorta. The endothelial dysfunction was prevented by the p38 antagonist SB203580, but not by the ERK1/2 antagonist PD98059 or JNK antagonist SP600125. Furthermore, the expression of p-eNOS, HIF-1α, and HIF-2α increased while that of p-p38 and BMP-4 decreased in CIHH-treated aortas from 2K1C rats. Finally, the p-eNOS expression was upregulated and the p-p38 expression was downregulated by pre-incubation of SB203580 or the BMP-4 antagonist Noggin with the aorta. CIHH ameliorated the impairment of endothelium-dependent relaxation through upregulating the expression of p-eNOS, which may be mediated by the inhibition of BMP-4/p-p38 MAPK, and upregulating the expression of HIFs in the 2K1C rat aorta.


Asunto(s)
Aorta/patología , Hipertensión/patología , Hipoxia/patología , Riñón/patología , Instrumentos Quirúrgicos , Acetilcolina/farmacología , Animales , Antracenos/farmacología , Antracenos/uso terapéutico , Aorta/efectos de los fármacos , Aorta/fisiopatología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Presión Sanguínea/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Proteína Morfogenética Ósea 4/metabolismo , Proteínas Portadoras/farmacología , Enfermedad Crónica , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Flavonoides/farmacología , Flavonoides/uso terapéutico , Hipertensión/tratamiento farmacológico , Hipertensión/fisiopatología , Hipoxia/fisiopatología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Imidazoles/farmacología , Imidazoles/uso terapéutico , Técnicas In Vitro , Masculino , Óxido Nítrico Sintasa de Tipo III/metabolismo , Nitroprusiato/farmacología , Fosforilación/efectos de los fármacos , Piridinas/farmacología , Piridinas/uso terapéutico , Ratas Sprague-Dawley , Sístole , Vasodilatación/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
J Physiol ; 595(14): 4973-4989, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28488367

RESUMEN

KEY POINTS: Central hypercapnic hypoventilation is highly prevalent in children suffering from congenital central hypoventilation syndrome (CCHS). Mutations of the gene for paired-like homeobox 2b (Phox2b) are aetiologically associated with CCHS and Phox2b is present in central components of respiratory chemoreflex, such as the nucleus tractus solitarius (NTS). Injection of the neurotoxin substance P-saporin into NTS destroys Phox2b-expressing neurons. Impaired hypercapnic ventilatory response caused by this neurotoxin is attributable to a loss of CO2 -sensitive Phox2b-expressing NTS neurons. A subgroup of Phox2b-expressing neurons exhibits intrinsic chemosensitivity. A background K+ channel-like current is partially responsible for such chemosensitivity in Phox2b-expressing neurons. The present study helps us better understand the mechanism of respiratory deficits in CCHS and potentially locates a brainstem site for development of precise clinical intervention. ABSTRACT: The nucleus tractus solitarius (NTS) neurons have been considered to function as central respiratory chemoreceptors. However, the common molecular marker defined for these neurons remains unknown. The present study investigated whether paired-like homeobox 2b (Phox2b)-expressing NTS neurons are recruited in hypercapnic ventilatory response (HCVR) and whether these neurons exhibit intrinsic chemosensitivity. HCVR was assessed using whole body plethysmography and neuronal chemosensitivity was examined by patch clamp recordings in brainstem slices or dissociated neurons from Phox2b-EGFP transgenic mice. Injection of the neurotoxin substance P-saporin (SSP-SAP) into NTS destroyed Phox2b-expressing neurons. Minute ventilation and tidal volume were both reduced by 13% during exposure to 8% CO2 in inspired air when ∼13% of the Phox2b-expressing neurons were eliminated. However, a loss of ∼18% of these neurons was associated with considerable decreases in minute ventilation by ≥18% and in tidal volume by≥22% when challenged by ≥4% CO2 . In both cases, breathing frequency was unaffected. Most CO2 -activated neurons were immunoreactive to Phox2b. In brainstem slices, ∼43% of Phox2b-expressing neurons from Phox2b-EGFP mice displayed a sustained or transient increase in firing rate during physiological acidification (pH 7.0 or 8% CO2 ). Such a response was also present in dissociated neurons in favour of an intrinsic property. In voltage clamp recordings, a background K+ channel-like current was found in a subgroup of Phox2b-expressing neurons. Thus, the respiratory deficits caused by injection of SSP-SAP into the NTS are attributable to proportional lesions of CO2 /H+ -sensitive Phox2b-expressing neurons.


Asunto(s)
Proteínas de Homeodominio/fisiología , Hipercapnia/fisiopatología , Neuronas/fisiología , Núcleo Solitario/fisiología , Factores de Transcripción/fisiología , Animales , Proteínas de Homeodominio/genética , Masculino , Potenciales de la Membrana , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neurotoxinas/toxicidad , Respiración , Proteínas Inactivadoras de Ribosomas Tipo 1/toxicidad , Saporinas , Núcleo Solitario/efectos de los fármacos , Factores de Transcripción/genética
9.
Neurosci Lett ; 635: 8-16, 2016 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-27760384

RESUMEN

OBJECTIVE: Providing adequate protection against cerebral ischemia remains an unrealized goal. The present study was aimed at testing whether chronic intermittent hypobaric hypoxia (CIHH) would have protective effects against cerebral ischemia and investigating the potential role of mitochondrial membrane ATP-sensitive potassium channel (mitoKATP) in this effect. METHODS: Ischemia was induced in rats by occlusion of bilateral common carotid arteries for 8min on day 2 after bilateral vertebral arteries were permanently electrocauterized and CIHH was simulated in a hypoxic chamber. Learning and memory impairments were analyzed using the Morris water maze. The delay neuronal death (DND) in the hippocampus CA1 was observed by thionine staining. The expression of the two subunits of mitoKATP, SUR1 and Kir 6.2, and the concentration of cytochrome c (Cyt c) were observed by Western blotting. The mitochondrial membrane potential (Δym) was determined by flow cytometry. Morphological changes of the mitochondria were investigated by electron microscopy. The antagonist of mitoKATP, 5-hydroxydecanoate (5-HD), was used to demonstrate the involvement of mitoKATP. RESULTS: CIHH pretreatment ameliorated the learning and memory impairments produced by ischemia, concomitant with reduced DND in the hippocampus CA1 area. Expression levels of SUR1 and Kir6.2 both increased for at least one week after CIHH pretreatment. Levels of the two subunits were higher in the CIHH pretreatment combined with ischemia group than the ischemia only group at 2 d and 7 d after ischemia. Furthermore, the concentration of Cyt c was decreased in mitochondria and increased in the cytoplasm after ischemia which was prevented by CIHH. The decrease of Δψm and the destruction of mitochondrial ultrastructure were both rescued by CIHH pretreatment. The above protective effects of CIHH were blocked by 5-HD intraperitoneal injection 30min before ischemia. CONCLUSION: CIHH pretreatment can reduce cerebral ischemic injury, which is mediated by upregulating the expression and activity of mitoKATP.


Asunto(s)
Isquemia Encefálica/prevención & control , Hipoxia , Canales de Potasio/metabolismo , Animales , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Isquemia Encefálica/psicología , Región CA1 Hipocampal/patología , Citocromos c/metabolismo , Masculino , Aprendizaje por Laberinto , Potencial de la Membrana Mitocondrial , Trastornos de la Memoria/prevención & control , Mitocondrias/patología , Mitocondrias/fisiología , Canales de Potasio de Rectificación Interna/metabolismo , Presión , Células Piramidales/patología , Ratas Wistar , Memoria Espacial , Receptores de Sulfonilureas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...