Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Lung Cell Mol Physiol ; 324(3): L259-L270, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36692168

RESUMO

Severe levels of acidosis (pH < 6.8) have been shown to cause a sustained rise in cytosolic Ca2+ concentration in carotid body Type 1 (glomus) cells. To understand how physiologically relevant levels of acidosis regulate Ca2+ signaling in glomus cells, we studied the effects of small changes in extracellular pH (pHo) on the kinetics of Ca2+ oscillations. A decrease in pHo from 7.4 to 7.3 (designated mild) and 7.2 (designated moderate) acidosis produced significant increases in the frequency and amplitude of Ca2+ oscillations. These effects of acidosis on Ca2+ oscillations were not blocked by NS383 and amiloride [acid-sensing ion channel (ASIC) inhibitors]. Mild and moderate levels of acidosis, however, caused a small but significant inhibition of two-pore domain acid-sensing K+ channels (TASK) (TASK-1- and TASK-3-like channels) and depolarized the cell by 6-13 mV. Acidosis-induced increase in Ca2+ oscillations was inhibited by nifedipine (1 µM; L-type Cav inhibitor) and by TTA-P2 (20 µM; T-type Cav inhibitor). Mild inhibition of TASK activity by N-[(2,4-difluorophenyl)methyl]-2'-[[[2-(4methoxyphenyl)acetyl]amino]methyl][1,1'-biphenyl]-2-carboxamide (A1899) (0.3 µM) and 1-[1-[6-[[1,1'-biphenyl]-4-ylcarbonyl)-5,6,7,8-tetrahydropyrido[4,3-d]pyrimidine-4-yl]-4-piperidinyl]-1-butanon (PK-THPP) (0.1 µM) increased Ca2+ oscillation frequency to levels similar to those observed with mild-moderate acidosis. Mild acidosis (pHo 7.3) and mild hypoxia (∼5%O2) produced similar levels of changes in the kinetics of Ca2+ oscillations. Block of tetraethylammonium (TEA)-sensitive Kv channels did not affect acid-induced increase in Ca2+ oscillations. Our study shows that mild and moderate levels of acidosis increase the frequency and amplitude of Ca2+ oscillations primarily by inhibition of TASK without involving ASICs, and suggests a major role of TASK for signal transduction in response to a physiological change in pHo.


Assuntos
Acidose , Corpo Carotídeo , Ratos , Animais , Células Quimiorreceptoras , Ácidos , Concentração de Íons de Hidrogênio , Cálcio
2.
Cell Tissue Res ; 384(2): 255-273, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33852077

RESUMO

Oxygen-chemoreceptive cells play critical roles for the respiration control. This review summarizes the chemoreceptive cells in the carotid body and fish gills from a morphological and molecular perspective. The cells synthesize and secrete biogenic amines, neuropeptides, and neuroproteins and also express many signaling molecules and transcription factors. In mammals, birds, reptiles, and amphibians, the carotid body primordium is consistently formed in the wall of the third arch artery which gives rise to the common carotid artery and the basal portion of the internal carotid artery. Consequently, the carotid body is located in the carotid bifurcation region, except birds in which the organ is situated at the lateral side of the common carotid artery. The carotid body receives branches of the cranial nerves IX and/or X dependent on the location of the organ. The glomus cell progenitors in mammals and birds are derived from the neighboring ganglion, i.e., the superior cervical sympathetic ganglion and the nodose ganglion, respectively, and immigrate into the carotid body primordium, constituting a solid cell cluster. In other animal species, the glomus cells are dispersed singly or forming small cell groups in intervascular stroma of the carotid body. In fishes, the neuroepithelial cells, corresponding to the glomus cells, are distributed in the gill filaments and lamellae. All oxygen-chemoreceptive cells sensitively respond to acute or chronic hypoxia, exhibiting degranulation, hypertrophy, hyperplasia, and upregulated expression of many genes.


Assuntos
Corpo Carotídeo/metabolismo , Células Quimiorreceptoras/metabolismo , Oxigênio/metabolismo , Animais , Peixes
3.
Dev Biol ; 444 Suppl 1: S308-S324, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29807017

RESUMO

Carotid body glomus cells mediate essential reflex responses to arterial blood hypoxia. They are dopaminergic and secrete growth factors that support dopaminergic neurons, making the carotid body a potential source of patient-specific cells for Parkinson's disease therapy. Like adrenal chromaffin cells, which are also hypoxia-sensitive, glomus cells are neural crest-derived and require the transcription factors Ascl1 and Phox2b; otherwise, their development is little understood at the molecular level. Here, analysis in chicken and mouse reveals further striking molecular parallels, though also some differences, between glomus and adrenal chromaffin cell development. Moreover, histology has long suggested that glomus cell precursors are 'émigrés' from neighbouring ganglia/nerves, while multipotent nerve-associated glial cells are now known to make a significant contribution to the adrenal chromaffin cell population in the mouse. We present conditional genetic lineage-tracing data from mice supporting the hypothesis that progenitors expressing the glial marker proteolipid protein 1, presumably located in adjacent ganglia/nerves, also contribute to glomus cells. Finally, we resolve a paradox for the 'émigré' hypothesis in the chicken - where the nearest ganglion to the carotid body is the nodose, in which the satellite glia are neural crest-derived, but the neurons are almost entirely placode-derived - by fate-mapping putative nodose neuronal 'émigrés' to the neural crest.


Assuntos
Corpo Carotídeo/embriologia , Células Cromafins/metabolismo , Pericitos/metabolismo , Glândulas Suprarrenais/metabolismo , Glândulas Suprarrenais/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Padronização Corporal/fisiologia , Diferenciação Celular , Hipóxia Celular/fisiologia , Embrião de Galinha , Galinhas/metabolismo , Camundongos , Camundongos Knockout , Proteína Proteolipídica de Mielina/fisiologia , Crista Neural/metabolismo , Neurônios/metabolismo , Pericitos/fisiologia , Fatores de Transcrição/metabolismo
4.
Neuroscience ; 310: 430-41, 2015 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-26431621

RESUMO

Angiotensin II (ANG II) is known to promote leptin production and secretion. Although ANG II type 1 receptors (AT1Rs) and leptin are expressed within the carotid body, it is not known whether AT1R and leptin are co-expressed in the same glomus cells nor if these peptides are affected within the carotid body by intermittent hypoxia (IH). This study was done to investigate whether ANG II modulated leptin signaling in the carotid body during IH. Rats were treated with captopril (Capt) or the AT1R blocker losartan (Los) in the drinking water for 3days prior to being exposed to IH (8h) or normoxia (8h). IH induced increases in plasma ANG II and leptin compared to normoxic controls. Capt treatment abolished the plasma leptin changes to IH, whereas Los treatment had no effect on the IH induced increase in plasma leptin. Additionally, carotid body glomus cells containing both leptin and the long form of the leptin receptor (OB-Rb) were found to co-express AT1R protein, and IH increased the expression of only AT1R protein within the carotid body in both Capt- and non-Capt-treated animals. On the other hand, Los treatment did not modify AT1R protein expression to IH. Additionally, Capt and Los treatment eliminated the elevated carotid body leptin protein expression, and the changes in phosphorylated signal transducer and activator of transcription three protein, the short form of the leptin receptor (OB-R100), suppressor of cytokine signaling 3, and phosphorylated extracellular-signal-regulated kinase 1/2 protein expression induced by IH. However, Capt elevated the expression of OB-Rb protein, whereas Los abolished the changes in OB-Rb protein to IH. These findings, taken together with the previous observation that ANG II modifies carotid body chemosensitivity, suggest that the increased circulating levels of ANG II and leptin induced by IH act at the carotid body to alter leptin signaling within the carotid body which in turn may influence chemoreceptor function.


Assuntos
Corpo Carotídeo/metabolismo , Hipóxia Encefálica/metabolismo , Leptina/metabolismo , Receptor Tipo 2 de Angiotensina/metabolismo , Angiotensina II/sangue , Bloqueadores do Receptor Tipo 1 de Angiotensina II/administração & dosagem , Bloqueadores do Receptor Tipo 2 de Angiotensina II/metabolismo , Animais , Captopril/administração & dosagem , Corpo Carotídeo/efeitos dos fármacos , Leptina/sangue , Losartan/administração & dosagem , Masculino , Ratos , Ratos Sprague-Dawley , Receptores para Leptina/metabolismo , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA