Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 640
Filter
1.
Int J Mol Sci ; 21(21)2020 Nov 03.
Article in English | MEDLINE | ID: mdl-33153142

ABSTRACT

The carotid body (CB), a neural-crest-derived organ and the main arterial chemoreceptor in mammals, is composed of clusters of cells called glomeruli. Each glomerulus contains neuron-like, O2-sensing glomus cells, which are innervated by sensory fibers of the petrosal ganglion and are located in close contact with a dense network of fenestrated capillaries. In response to hypoxia, glomus cells release transmitters to activate afferent fibers impinging on the respiratory and autonomic centers to induce hyperventilation and sympathetic activation. Glomus cells are embraced by interdigitating processes of sustentacular, glia-like, type II cells. The CB has an extraordinary structural plasticity, unusual for a neural tissue, as it can grow several folds its size in subjects exposed to sustained hypoxia (as for example in high altitude dwellers or in patients with cardiopulmonary diseases). CB growth in hypoxia is mainly due to the generation of new glomeruli and blood vessels. In recent years it has been shown that the adult CB contains a collection of quiescent multipotent stem cells, as well as immature progenitors committed to the neurogenic or the angiogenic lineages. Herein, we review the main properties of the different cell types in the CB germinal niche. We also summarize experimental data suggesting that O2-sensitive glomus cells are the master regulators of CB plasticity. Upon exposure to hypoxia, neurotransmitters and neuromodulators released by glomus cells act as paracrine signals that induce proliferation and differentiation of multipotent stem cells and progenitors, thus causing CB hypertrophy and an increased sensory output. Pharmacological modulation of glomus cell activity might constitute a useful clinical tool to fight pathologies associated with exaggerated sympathetic outflow due to CB overactivation.


Subject(s)
Carotid Body/cytology , Neurotransmitter Agents/physiology , Stem Cell Niche/physiology , Adaptation, Physiological/physiology , Animals , Cell Differentiation/physiology , Humans , Hypoxia/metabolism , Hypoxia/physiopathology , Neurogenesis/physiology , Neurons/physiology , Neurotransmitter Agents/metabolism , Oxygen/metabolism
2.
Int J Mol Sci ; 21(19)2020 10 01.
Article in English | MEDLINE | ID: mdl-33019660

ABSTRACT

The carotid body may undergo plasticity changes during development/ageing and in response to environmental (hypoxia and hyperoxia), metabolic, and inflammatory stimuli. The different cell types of the carotid body express a wide series of growth factors and corresponding receptors, which play a role in the modulation of carotid body function and plasticity. In particular, type I cells express nerve growth factor, brain-derived neurotrophic factor, neurotrophin 3, glial cell line-derived neurotrophic factor, ciliary neurotrophic factor, insulin-like-growth factor-I and -II, basic fibroblast growth factor, epidermal growth factor, transforming growth factor-α and -ß, interleukin-1ß and -6, tumor necrosis factor-α, vascular endothelial growth factor, and endothelin-1. Many specific growth factor receptors have been identified in type I cells, indicating autocrine/paracrine effects. Type II cells may also produce growth factors and express corresponding receptors. Future research will have to consider growth factors in further experimental models of cardiovascular, metabolic, and inflammatory diseases and in human (normal and pathologic) samples. From a methodological point of view, microarray and/or proteomic approaches would permit contemporary analyses of large groups of growth factors. The eventual identification of physical interactions between receptors of different growth factors and/or neuromodulators could also add insights regarding functional interactions between different trophic mechanisms.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Carotid Body/metabolism , Hyperoxia/genetics , Hypoxia/genetics , Nerve Growth Factor/genetics , Receptors, Growth Factor/genetics , Animals , Brain-Derived Neurotrophic Factor/metabolism , Carotid Body/cytology , Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor/metabolism , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Hyperoxia/metabolism , Hyperoxia/pathology , Hypoxia/metabolism , Hypoxia/pathology , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Nerve Growth Factor/metabolism , Neurotrophin 3/genetics , Neurotrophin 3/metabolism , Receptors, Growth Factor/metabolism , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
3.
J Histochem Cytochem ; 68(10): 679-690, 2020 10.
Article in English | MEDLINE | ID: mdl-32886017

ABSTRACT

TWIK-related acid-sensitive K+ (TASK) homomeric channels, TASK1 and TASK3, are present in PC12 cells. The channels do not heteromerize due plausibly to a lack of p11 protein. Single-channel recording reveals that most of the rat carotid body (CB) glomus cells express heteromeric TASK1-TASK3 channels, but the presence of p11 in glomus cells has not yet been verified. TASK1, but not TASK3, binds to p11, which has a retention signal for the endoplasmic reticulum. We hypothesized that p11 could facilitate heteromeric TASK1-TASK3 formation in glomus cells. We investigated this hypothesis in isolated immunocytochemically identified rat CB glomus cells. The findings were that glomus cells expressed p11-like immunoreactive (IR) material, and TASK1- and TASK3-like IR material mainly coincided in the cytoplasm. The proximity ligation assay showed that TASK1 and TASK3 heteromerized. In separate experiments, supporting evidence for the major role of p11 for channel heteromerization was provided in PC12 cells stimulated by nerve growth factor. p11 production took place there via multiple signaling pathways comprising mitogen-activated protein kinase and phospholipase C, and heteromeric TASK1-TASK3 channels were formed. We conclude that p11 is expressed and TASK1 and TASK3 heteromerize in rat CB glomus cells.


Subject(s)
Annexin A2/biosynthesis , Carotid Body/metabolism , Nerve Tissue Proteins/biosynthesis , Potassium Channels, Tandem Pore Domain/biosynthesis , S100 Proteins/biosynthesis , Animals , Carotid Body/cytology , Immunohistochemistry , Male , PC12 Cells , Rats , Rats, Wistar
4.
Acta Histochem ; 122(3): 151507, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31955909

ABSTRACT

Vesicular monoamine transporters (VMAT) 1 and 2 are responsible for monoamine transportation into secretary vesicles and are tissue-specifically expressed in central and peripheral monoaminergic tissues, including the carotid body (CB). The aim of the present study was to examine the expression of catecholamine-synthesizing enzymes in VMAT1- and VMAT2-immunoreactive glomus cells in the rat CB using multiple immunolabeling. The expression of VMAT1 and VMAT2 mRNA in the CB was confirmed by RT-PCR. Immunohistochemistry revealed that VMAT1 immunoreactivity was predominant in glomus cells rather than VMAT2 immunoreactivity. Glomus cells with VMAT1 immunoreactivity exhibited weak/negative VMAT2 immunoreactivity, and vice versa. Immunoreactivities for VMAT1 and tyrosine hydroxylase, the rate-limiting enzyme for catecholamine biosynthesis, were co-localized in the same glomus cells and a positive correlation was confirmed between the two immunoreactivities (Spearman's coefficient = 0.82; p <  0.05). Although some glomus cells showed co-localization of VMAT2 and dopamine ß-hydroxylase immunoreactivity, the biosynthetic enzyme for noradrenaline, VMAT2 immunoreactivity appeared to be less associated with both catecholamine-synthesizing enzymes as indicated by a correlation analysis (TH: Spearman's coefficient = 0.38, DBH: Spearman's coefficient = 0.26). These results indicate that heterogeneity on functional role would exist among glomus cells in terms of VMAT isoform and catecholamine-synthesizing enzymes expression.


Subject(s)
Carotid Body/metabolism , Catecholamines/biosynthesis , Vesicular Monoamine Transport Proteins/metabolism , Animals , Carotid Body/cytology , Dopamine beta-Hydroxylase/metabolism , Immunohistochemistry , Male , Norepinephrine/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Vesicular Monoamine Transport Proteins/genetics
5.
Sci Signal ; 13(615)2020 01 21.
Article in English | MEDLINE | ID: mdl-31848220

ABSTRACT

Acute cardiorespiratory responses to O2 deficiency are essential for physiological homeostasis. The prototypical acute O2-sensing organ is the carotid body, which contains glomus cells expressing K+ channels whose inhibition by hypoxia leads to transmitter release and activation of nerve fibers terminating in the brainstem respiratory center. The mechanism by which changes in O2 tension modulate ion channels has remained elusive. Glomus cells express genes encoding HIF2α (Epas1) and atypical mitochondrial subunits at high levels, and mitochondrial NADH and reactive oxygen species (ROS) accumulation during hypoxia provides the signal that regulates ion channels. We report that inactivation of Epas1 in adult mice resulted in selective abolition of glomus cell responsiveness to acute hypoxia and the hypoxic ventilatory response. Epas1 deficiency led to the decreased expression of atypical mitochondrial subunits in the carotid body, and genetic deletion of Cox4i2 mimicked the defective hypoxic responses of Epas1-null mice. These findings provide a mechanistic explanation for the acute O2 regulation of breathing, reveal an unanticipated role of HIF2α, and link acute and chronic adaptive responses to hypoxia.


Subject(s)
Arteries/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chemoreceptor Cells/metabolism , Electron Transport Complex IV/metabolism , Animals , Arteries/cytology , Basic Helix-Loop-Helix Transcription Factors/genetics , Carotid Body/cytology , Carotid Body/metabolism , Electron Transport Complex IV/genetics , Hypoxia , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitochondria/metabolism , Oxygen/metabolism , Reactive Oxygen Species/metabolism , Respiratory System/metabolism , Signal Transduction
6.
J Physiol ; 597(19): 4991-5008, 2019 10.
Article in English | MEDLINE | ID: mdl-31426127

ABSTRACT

KEY POINTS: Adenosine and ATP are excitatory neurotransmitters involved in the carotid body (CB) response to hypoxia. During ageing the CB exhibits a decline in its functionality, demonstrated by decreased hypoxic responses. In aged rats (20-24 months old) there is a decrease in: basal and hypoxic release of adenosine and ATP from the CB; expression of adenosine and ATP receptors in the petrosal ganglion; carotid sinus nerve (CSN) activity in response to hypoxia; and ventilatory responses to ischaemic hypoxia. There is also an increase in SNAP25, ENT1 and CD73 expression. It is concluded that, although CSN activity and ventilatory responses to hypoxia decrease with age, adjustments in purinergic metabolism in the CB in aged animals are present aiming to maintain the contribution of adenosine and ATP. The possible significance of the findings in the context of ageing and in CB-associated pathologies is considered. ABSTRACT: During ageing the carotid body (CB) exhibits a decline in its functionality. Here we investigated the effect of ageing on functional CB characteristics as well as the contribution of adenosine and ATP to CB chemosensory activity. Experiments were performed in 3-month-old and 20- to 24-month-old male Wistar rats. Ageing decreased: the number of tyrosine hydroxylase immune-positive cells, but not type II cells or nestin-positive cells in the CB; the expression of P2X2 and A2A receptors in the petrosal ganglion; and the basal and hypoxic release of adenosine and ATP from the CB. Ageing increased ecto-nucleotidase (CD73) immune-positive cells and the expression of synaptosome associated protein 25 (SNAP25) and equilibrative nucleoside transporter 1 (ENT1) in the CB. Additionally, ageing did not modify basal carotid sinus nerve (CSN) activity or the activity in response to hypercapnia, but decreased CSN activity in hypoxia. The contribution of adenosine and ATP to stimuli-evoked CSN chemosensory activity in aged animals followed the same pattern of 3-month-old animals. Bilateral common carotid occlusions during 5, 10 and 15 s increased ventilation proportionally to the duration of ischaemia, an effect decreased by ageing. ATP contributed around 50% to ischaemic-ventilatory responses in young and aged rats; the contribution of adenosine was dependent on the intensity of ischaemia, being maximal in ischaemias of 5 s (50%) and much smaller in 15 s ischaemias. Our results demonstrate that both ATP and adenosine contribute to CB chemosensory activity in ageing. Though CB responses to hypoxia, but not to hypercapnia, decrease with age, the relative contribution of both ATP and adenosine for CB activity is maintained.


Subject(s)
Adenosine Triphosphate/metabolism , Adenosine/metabolism , Carotid Body/physiology , Chemoreceptor Cells/metabolism , Aging , Animals , Antinematodal Agents/pharmacology , Carotid Body/cytology , Female , Gene Expression Regulation/drug effects , Male , Rats , Rats, Wistar , Suramin/pharmacology , Triazines/pharmacology , Triazoles/pharmacology
7.
Adv Exp Med Biol ; 1123: 19-38, 2019.
Article in English | MEDLINE | ID: mdl-31016593

ABSTRACT

Somatic stem cells confer plasticity to adult tissues, permitting their maintenance, repair and adaptation to a changing environment. Adult germinal niches supporting somatic stem cells have been thoroughly characterized throughout the organism, including in central and peripheral nervous systems. Stem cells do not reside alone within their niches, but they are rather accompanied by multiple progenitor cells that not only contribute to the progression of stem cell lineage but also regulate their behavior. Understanding the mechanisms underlying these interactions within the niche is crucial to comprehend associated pathologies and to use stem cells in cell therapy. We have described a stunning germinal niche in the adult peripheral nervous system: the carotid body. This is a chemoreceptor organ with a crucial function during physiological adaptation to hypoxia. We have shown the presence of multipotent stem cells within this niche, escorted by multiple restricted progenitor cell types that contribute to niche physiology and hence organismal adaptation to the lack of oxygen. Herein, we discuss new and existing data about the nature of all these stem and progenitor cell types present in the carotid body germinal niche, discussing their role in physiology and their clinical relevance for the treatment of diverse pathologies.


Subject(s)
Carotid Body/cytology , Multipotent Stem Cells/cytology , Cell Lineage , Humans , Stem Cell Niche
8.
Respir Physiol Neurobiol ; 260: 17-27, 2019 02.
Article in English | MEDLINE | ID: mdl-30389452

ABSTRACT

In humans the intravenous anaesthetic propofol depresses ventilatory responses to hypoxia and CO2. Animal studies suggest that this may in part be due to inhibition of synaptic transmission between chemoreceptor glomus cells of the carotid body and the afferent carotid sinus nerve. It is however unknown if propofol can also act directly on the glomus cell. Here we report that propofol can indeed inhibit intracellular Ca2+ responses to hypoxia and hypercapnia in isolated rat glomus cells. Neither this propofol effect, nor the glomus cell response to hypoxia in the absence of propofol, were influenced by GABA receptor activation (using GABA, muscimol and baclofen) or inhibition (using bicuculline and 5-aminovaleric acid). Suggesting that these effects of propofol are not mediated through GABA receptors. Propofol inhibited calcium responses to nicotine in glomus cells but the nicotinic antagonists vecuronium and methyllycaconitine did not inhibit calcium responses to hypoxia. TASK channel activity was not altered by propofol. The glomus cell Ca2+ response to depolarisation with 30 mM K+ was however modestly inhibited by propofol. In summary we conclude that propofol does have a direct effect upon hypoxia signalling in isolated type-1 cells and that this may be partially due to its ability to inhibit voltage gated Ca2+v channels. We also note that propofol has the capacity to supress glomus cell excitation via nicotinic receptors and may therefore also interfere with paracrine/autocrine cholinergic signalling in the intact organ. The effects of propofol on chemoreceptor function are however clearly complex and require further investigation.


Subject(s)
Carotid Body/cytology , Cell Hypoxia/drug effects , Chemoreceptor Cells/drug effects , Hypercapnia/pathology , Hypnotics and Sedatives/pharmacology , Propofol/pharmacology , Animals , Animals, Newborn , Calcium/metabolism , Carbon Dioxide/pharmacology , Carotid Body/growth & development , Cholinergic Agents/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , GABA Agents/pharmacology , Membrane Potentials/drug effects , Patch-Clamp Techniques , Potassium/pharmacology , Rats , Rats, Sprague-Dawley
9.
Physiol Rep ; 6(19): e13876, 2018 09.
Article in English | MEDLINE | ID: mdl-30284397

ABSTRACT

Sensing of hypoxia and acidosis in arterial chemoreceptors is thought to be mediated through the inhibition of TASK and possibly other (e.g., BKCa ) potassium channels which leads to membrane depolarization, voltage-gated Ca-entry, and neurosecretion. Here, we investigate the effects of pharmacological inhibitors on TASK channel activity and [Ca2+ ]i -signaling in isolated neonatal rat type-1 cells. PK-THPP inhibited TASK channel activity in cell attached patches by up to 90% (at 400 nmol/L). A1899 inhibited TASK channel activity by 35% at 400 nmol/L. PK-THPP, A1899 and Ml 365 all evoked a rapid increase in type-1 cell [Ca2+ ]i . These [Ca2+ ]i responses were abolished in Ca2+ -free solution and greatly attenuated by Ni2+ (2 mM) suggesting that depolarization and voltage-gated Ca2+ -entry mediated the rise in [Ca2+ ]i. Doxapram (50 µmol/L), a respiratory stimulant, also inhibited type-1 cell TASK channel activity and increased [Ca2+ ]i. . We also tested the effects of combined inhibition of BKCa and TASK channels. TEA (5 mmol/L) slightly increased [Ca2+ ]i in the presence of PK-THPP and A1899. Paxilline (300 nM) and iberiotoxin (50 nmol/L) also slightly increased [Ca2+ ]i in the presence of A1899 but not in the presence of PK-THPP. In general [Ca2+ ]i responses to TASK inhibitors, alone or in combination with BKCa inhibitors, were smaller than the [Ca2+ ]i responses evoked by hypoxia. These data confirm that TASK channel inhibition is capable of evoking membrane depolarization and robust voltage-gated Ca2+ -entry but suggest that this, even with concomitant inhibition of BKCa channels, may be insufficient to account fully for the [Ca2+ ]i -response to hypoxia.


Subject(s)
Benzamides/pharmacology , Benzeneacetamides/pharmacology , Calcium Signaling/drug effects , Carotid Body/cytology , Carotid Body/drug effects , Doxapram/pharmacology , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Animals , Animals, Newborn , Calcium Signaling/physiology , Carotid Body/physiology , HEK293 Cells , Humans , Nerve Tissue Proteins , Potassium Channels, Tandem Pore Domain/metabolism , Rats , Rats, Sprague-Dawley , Respiratory System Agents/pharmacology
10.
Adv Exp Med Biol ; 1071: 35-41, 2018.
Article in English | MEDLINE | ID: mdl-30357731

ABSTRACT

Glomus cells isolated from rabbit and rat/mouse carotid bodies have been used for many years to study the role of ion channels in hypoxia sensing. Studies show that hypoxia inhibits the inactivating K+ channels (Kv4) in rabbits, but inhibits TASK in rats/mice to elicit the hypoxic response. Because the role of TASK in rabbit glomus cells is not known, we isolated glomus cells from rabbits and studied the expression of TASK mRNA in the whole carotid body (CB), changes in [Ca2+]i and TASK activity. RT-PCR showed that rabbit CB expressed mRNA for TASK-3 and several Kv (Kv2.1, Kv3.1 and Kv3.3). In rabbit glomus cells in which 20 mM KClo elevated [Ca2+], anoxia also elicited a strong rise in [Ca2+]. In cell-attached patches with 140 mM KCl in the pipette, basal openings of ion channels with single-channel conductance levels of 16-pS, 34-pS, and 42-pS were present. TREK-like channels were also observed. In inside-out patches with high [Ca2+]i, BK was activated. The 42-pS channel opened spontaneously and briefly. The 16-pS and 34-pS channels showed properties similar to those of TASK-1 and TASK-3, respectively. TASK activity in cell-attached patches was lower than that in rat glomus cells under identical recording conditions. Hypoxia (~0.5%O2) reduced TASK activity by ~52% and depolarized the cells by ~30 mV. Our results show that the O2-sensitive TASK contributes to the hypoxic response in rabbit glomus cells.


Subject(s)
Carotid Body/cytology , Potassium Channels, Tandem Pore Domain/physiology , Animals , Calcium/physiology , Cell Hypoxia , Membrane Potentials , Mice , Nerve Tissue Proteins , Patch-Clamp Techniques , Rabbits , Rats
11.
Physiol Genomics ; 50(7): 504-509, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29652633

ABSTRACT

Known primarily for its oxygen-sensing capabilities, the carotid body chemoreceptors have recently been implicated, primarily by work in animal models, in the pathophysiology of a number of metabolic conditions. The research presented in this brief review highlights translational work conducted at the Mayo Clinic between 2010 and 2017 in healthy humans and discusses key areas for future work in disease populations.


Subject(s)
Carotid Body/metabolism , Chemoreceptor Cells/metabolism , Glucose/metabolism , Insulin/metabolism , Animals , Carotid Body/cytology , Humans , Hypoxia , Oxygen/metabolism , Translational Research, Biomedical/methods , Translational Research, Biomedical/trends
12.
Methods Mol Biol ; 1742: 125-137, 2018.
Article in English | MEDLINE | ID: mdl-29330796

ABSTRACT

The carotid body is the main arterial chemoreceptor in mammals that mediates the cardiorespiratory reflexes activated by acute hypoxia. Here we describe the protocols followed in our laboratory to study responsiveness to hypoxia of single, enzymatically dispersed, glomus cells monitored by microfluorimetry and the patch-clamp technique.


Subject(s)
Carotid Body/cytology , Chemoreceptor Cells/metabolism , Patch-Clamp Techniques/methods , Single-Cell Analysis/methods , Animals , Carotid Body/physiology , Cell Hypoxia , Cells, Cultured , Cytophotometry , Mice , Rats
13.
Methods Mol Biol ; 1742: 139-153, 2018.
Article in English | MEDLINE | ID: mdl-29330797

ABSTRACT

Monitoring responsiveness to acute hypoxia of whole animals and single cells is essential to investigate the nature of the mechanisms underlying oxygen (O2) sensing. Here we describe the protocols followed in our laboratory to evaluate the ventilatory response to hypoxia in normal and genetically modified animals. We also describe the amperometric technique used to monitor single-cell catecholamine release from chemoreceptor cells in carotid body and adrenal medulla slices.


Subject(s)
Animals, Genetically Modified/metabolism , Catecholamines/metabolism , Chemoreceptor Cells/metabolism , Oxygen/metabolism , Adrenal Medulla/cytology , Adrenal Medulla/metabolism , Animals , Carotid Body/cytology , Carotid Body/metabolism , Cell Hypoxia , Mice , Plethysmography , Single-Cell Analysis
14.
Methods Mol Biol ; 1742: 155-166, 2018.
Article in English | MEDLINE | ID: mdl-29330798

ABSTRACT

Immunohistochemistry (IHC) enables the detection and distribution of proteins in cells of tissues. IHC has become an indispensable approach for studying oxygen sensing by the carotid body (CB). This chapter provides a detailed description of IHC of CB tissue and isolated CB cells.


Subject(s)
Carotid Body/cytology , Oxygen/metabolism , Animals , Carotid Body/metabolism , Immunohistochemistry , Mice , Microscopy, Fluorescence , Rats , Software
15.
J Physiol ; 596(15): 3119-3136, 2018 08.
Article in English | MEDLINE | ID: mdl-29160573

ABSTRACT

KEY POINTS: We studied the role of the large-conductance Ca2+ -activated K+ channel (BK) and voltage-dependent K+ channels (Kv) on [Ca2+ ]i responses to a wide range of hypoxia at different resting cell membrane potential (Em ). BK/Kv were mostly closed at rest in normoxia. BK/Kv became basally active when cells were depolarized by elevated [KCl]o (>12 mm). Regardless of whether BK/Kv were closed or basally open, hypoxia-induced elevation of [Ca2+ ]i was enhanced 2- to 3-fold by inhibitors of BK/Kv. Hypoxia-induced elevation of [Ca2+ ]i was enhanced ∼2-fold by an inhibitor of Kv2, a major Kv in rat glomus cells. Hypoxia did not inhibit BK in inside-out patches. Our study supports a scheme in which activation of BK/Kv strongly limits the magnitude of hypoxia-induced [Ca2+ ]i rise, with Kv having a much greater effect than BK. ABSTRACT: Large-conductance KCa (BK) and other voltage-dependent K+ channels (Kv) are highly expressed in carotid body (CB) glomus cells, but their role in hypoxia-induced excitation is still not well defined and remains controversial. We addressed this issue by studying the effects of inhibitors of BK (IBTX) and BK/Kv (TEA/4-AP) on [Ca2+ ]i responses to a wide range of hypoxia at different levels of resting cell membrane potential (Em ). IBTX and TEA/4-AP did not affect the basal [Ca2+ ]i in isolated glomus cells bathed in 5 mm KClo , but elicited transient increases in [Ca2+ ]i in cells that were moderately depolarized (11-20 mV) by elevation of [KCl]o (12-20 mm). Thus, BK and Kv were mostly closed at rest and activated by depolarization. Four different levels of hypoxia (mild, moderate, severe, anoxia) were used to produce a wide range of [Ca2+ ]i elevation (0-700 nm). IBTX did not affect the rise in [Ca2+ ]i , but TEA/4-AP strongly (∼3-fold) enhanced [Ca2+ ]i rise by moderate and severe levels of hypoxia. Guangxitoxin, a Kv2 blocker, inhibited the whole-cell current by ∼50%, and enhanced 2-fold the [Ca2+ ]i rise elicited by moderate and severe levels of hypoxia. Anoxia did not directly affect BK, but activated BK via depolarization. Our findings do not support the view that hypoxia inhibits BK/Kv to initiate or maintain the hypoxic response. Rather, our results show that BK/Kv are activated as glomus cells depolarize in response to hypoxia, which then limits the rise in [Ca2+ ]i . Inhibition of Kv may provide a mechanism to enhance the chemosensory activity of the CB and ventilation.


Subject(s)
Calcium/physiology , Carotid Body/physiology , Cell Hypoxia/physiology , Large-Conductance Calcium-Activated Potassium Channels/physiology , Potassium Channels, Voltage-Gated/physiology , Animals , Carotid Body/cytology , Rats, Sprague-Dawley
16.
Respir Physiol Neurobiol ; 246: 92-97, 2017 12.
Article in English | MEDLINE | ID: mdl-28782663

ABSTRACT

Respiratory activity is controlled by inputs from the peripheral and central chemoreceptors. Since overactivity of the carotid bodies, the main peripheral chemoreceptors, is linked to the pathophysiology of disparate metabolic and cardiovascular diseases, carotid body denervation (CBD) has been proposed as a potential treatment. However, long-term effects of CBD on the respiratory rhythm and regularity of breathing remain unknown. Here, we show that five weeks after bilateral CBD in rats, the respiratory rhythm was slower and less regular. Ten weeks after bilateral CBD, the respiratory frequency was not different from the sham-operated group, but the regularity of the respiratory rhythm was still reduced. Increased frequency of randomly occurring apneas is likely to be responsible for the irregular breathing pattern after CBD. These results should be taken into consideration since any treatment that reduces the stability of the respiratory rhythm might exacerbate the cardio-respiratory instability and worsen the cardiovascular outcomes.


Subject(s)
Carotid Body/physiopathology , Respiration Disorders/etiology , Respiration , Respiratory Mechanics/physiology , Animals , Carotid Body/cytology , Chemoreceptor Cells/physiology , Denervation/adverse effects , Male , Plethysmography , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric
17.
J Appl Physiol (1985) ; 123(5): 1335-1343, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28819004

ABSTRACT

Maintaining oxygen homeostasis is crucial to the survival of animals. Mammals respond acutely to changes in blood oxygen levels by modulating cardiopulmonary function. The major sensor of blood oxygen that regulates breathing is the carotid body (CB), a small chemosensory organ located at the carotid bifurcation. When arterial blood oxygen levels drop in hypoxia, neuroendocrine cells in the CB called glomus cells are activated to signal to afferent nerves that project to the brain stem. The mechanism by which hypoxia stimulates CB sensory activity has been the subject of many studies over the past 90 years. Two discrete models emerged that argue for the seat of oxygen sensing to lie either in the plasma membrane or mitochondria of CB cells. Recent studies are bridging the gap between these models by identifying hypoxic signals generated by changes in mitochondrial function in the CB that can be sensed by plasma membrane proteins on glomus cells. The CB is important for physiological adaptation to hypoxia, and its dysfunction contributes to sympathetic hyperactivity in common conditions such as sleep-disordered breathing, chronic heart failure, and insulin resistance. Understanding the basic mechanism of oxygen sensing in the CB could allow us to develop strategies to target this organ for therapy. In this short review, I will describe two historical models of CB oxygen sensing and new findings that are integrating these models.


Subject(s)
Carotid Body/physiology , Cell Membrane/metabolism , Mitochondria/metabolism , Oxygen/metabolism , Adaptation, Physiological , Animals , Carotid Body/cytology , Humans , Respiration
18.
J Physiol ; 595(18): 6091-6120, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28718507

ABSTRACT

KEY POINTS: Glomus cells in the carotid body (CB) and chromaffin cells in the adrenal medulla (AM) are essential for reflex cardiorespiratory adaptation to hypoxia. However, the mechanisms whereby these cells detect changes in O2 tension are poorly understood. The metabolic properties of acute O2 -sensing cells have been investigated by comparing the transcriptomes of CB and AM cells, which are O2 -sensitive, with superior cervical ganglion neurons, which are practically O2 -insensitive. In O2 -sensitive cells, we found a characteristic prolyl hydroxylase 3 down-regulation and hypoxia inducible factor 2α up-regulation, as well as overexpression of genes coding for three atypical mitochondrial electron transport subunits and pyruvate carboxylase, an enzyme that replenishes tricarboxylic acid cycle intermediates. In agreement with this observation, the inhibition of succinate dehydrogenase impairs CB acute O2 sensing. The responsiveness of peripheral chemoreceptor cells to acute hypoxia depends on a 'signature metabolic profile'. ABSTRACT: Acute O2 sensing is a fundamental property of cells in the peripheral chemoreceptors, e.g. glomus cells in the carotid body (CB) and chromaffin cells in the adrenal medulla (AM), and is necessary for adaptation to hypoxia. These cells contain O2 -sensitive ion channels, which mediate membrane depolarization and transmitter release upon exposure to hypoxia. However, the mechanisms underlying the detection of changes in O2 tension by cells are still poorly understood. Recently, we suggested that CB glomus cells have specific metabolic features that favour the accumulation of reduced quinone and the production of mitochondrial NADH and reactive oxygen species during hypoxia. These signals alter membrane ion channel activity. To investigate the metabolic profile characteristic of acute O2 -sensing cells, we used adult mice to compare the transcriptomes of three cell types derived from common sympathoadrenal progenitors, but exhibiting variable responsiveness to acute hypoxia: CB and AM cells, which are O2 -sensitive (glomus cells > chromaffin cells), and superior cervical ganglion neurons, which are practically O2 -insensitive. In the O2 -sensitive cells, we found a characteristic mRNA expression pattern of prolyl hydroxylase 3/hypoxia inducible factor 2α and up-regulation of several genes, in particular three atypical mitochondrial electron transport subunits and some ion channels. In addition, we found that pyruvate carboxylase, an enzyme fundamental to tricarboxylic acid cycle anaplerosis, is overexpressed in CB glomus cells. We also observed that the inhibition of succinate dehydrogenase impairs CB acute O2 sensing. Our data suggest that responsiveness to acute hypoxia depends on a 'signature metabolic profile' in chemoreceptor cells.


Subject(s)
Carotid Body/cytology , Chemoreceptor Cells/metabolism , Hypoxia/metabolism , Oxygen/metabolism , Transcriptome , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carotid Body/metabolism , Cells, Cultured , Female , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Male , Mice , Mice, Inbred C57BL , Potassium Channels/genetics , Potassium Channels/metabolism , Procollagen-Proline Dioxygenase/genetics , Procollagen-Proline Dioxygenase/metabolism
19.
Cell Rep ; 19(3): 471-478, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28423311

ABSTRACT

Adult stem cell plasticity, or the ability of somatic stem cells to cross boundaries and differentiate into unrelated cell types, has been a matter of debate in the last decade. Neural-crest-derived stem cells (NCSCs) display a remarkable plasticity during development. Whether adult populations of NCSCs retain this plasticity is largely unknown. Herein, we describe that neural-crest-derived adult carotid body stem cells (CBSCs) are able to undergo endothelial differentiation in addition to their reported role in neurogenesis, contributing to both neurogenic and angiogenic processes taking place in the organ during acclimatization to hypoxia. Moreover, CBSC conversion into vascular cell types is hypoxia inducible factor (HIF) dependent and sensitive to hypoxia-released vascular cytokines such as erythropoietin. Our data highlight a remarkable physiological plasticity in an adult population of tissue-specific stem cells and could have impact on the use of these cells for cell therapy.


Subject(s)
Adult Stem Cells/physiology , Carotid Body/cytology , Mammals/metabolism , Neural Stem Cells/physiology , Neuronal Plasticity , Adult Stem Cells/cytology , Adult Stem Cells/drug effects , Animals , Blood Vessels/cytology , Cell Differentiation/drug effects , Cell Hypoxia/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Erythropoietin/pharmacology , Female , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice, Transgenic , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Neovascularization, Physiologic/drug effects , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Neuronal Plasticity/drug effects
20.
J Physiol ; 595(13): 4261-4277, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28332205

ABSTRACT

KEY POINTS: 5-HT is a neuromodulator released from carotid body (CB) chemoreceptor (type I) cells and facilitates the sensory discharge following chronic intermittent hypoxia (CIH). In the present study, we show that, in addition to type I cells, adjacent glial-like type II cells express functional, ketanserin-sensitive 5-HT2 receptors, and their stimulation increases cytoplasmic Ca2+ derived from intracellular stores. In type II cells, 5-HT activated a ketanserin-sensitive inward current (I5-HT ) that was similar to that (IUTP ) activated by the P2Y2R agonist, UTP. As previously shown for IUTP , I5-HT was inhibited by BAPTA-AM and carbenoxolone (5 µm), a putative blocker of ATP-permeable pannexin (Panx)-1 channels; IUTP was reversibly inhibited by the specific Panx-1 mimetic peptide channel blocker, 10 Panx peptide. Paracrine stimulation of type II cells by 5-HT, leading to ATP release via Panx-1 channels, may contribute to CB excitability, especially in pathophysiological conditions associated with CIH (e.g. obstructive sleep apnoea). ABSTRACT: Carotid body (CB) chemoreceptor (type I) cells can synthesize and release 5-HT and increased autocrine-paracrine 5-HT2 receptor signalling contributes to sensory long-term facilitation during chronic intermittent hypoxia (CIH). However, recent studies suggest that adjacent glial-like type II cells can respond to CB paracrine signals by elevating intracellular calcium (Δ[Ca2+ ]i ) and activating carbenoxolone-sensitive, ATP-permeable, pannexin (Panx)-1-like channels. In the present study, using dissociated rat CB cultures, we found that 5-HT induced Δ[Ca2+ ]i responses in a subpopulation of type I cells, as well as in most (∼67%) type II cells identified by their sensitivity to the P2Y2 receptor agonist, UTP. The 5-HT-induced Ca2+ response in type II cells was dose-dependent (EC50 ∼183 nm) and largely inhibited by the 5-HT2A receptor blocker, ketanserin (1 µm), and also arose mainly from intracellular stores. 5-HT also activated an inward current (I5-HT ) in type II cells (EC50 ∼200 nm) that was reversibly inhibited by ketanserin (1-10 nm), the Ca2+ chelator BAPTA-AM (5 µm), and low concentrations of carbenoxolone (5 µm), a putative Panx-1 channel blocker. I5-HT reversed direction at approximately -11 mV and was indistinguishable from the UTP-activated current (IUTP ). Consistent with a role for Panx-1 channels, IUTP was reversibly inhibited by the specific Panx-1 mimetic peptide blocker 10 Panx (100 µm), although not by its scrambled control peptide (sc Panx). Because ATP is an excitatory CB neurotransmitter, it is possible that the contribution of enhanced 5-HT signalling to the increased sensory discharge during CIH may occur, in part, by a boosting of ATP release from type II cells via Panx-1 channels.


Subject(s)
Action Potentials , Calcium Signaling , Carotid Body/metabolism , Chemoreceptor Cells/metabolism , Connexins/metabolism , Nerve Tissue Proteins/metabolism , Serotonin Receptor Agonists/pharmacology , Serotonin/pharmacology , Animals , Carbenoxolone/pharmacology , Carotid Body/cytology , Cells, Cultured , Chemoreceptor Cells/drug effects , Connexins/antagonists & inhibitors , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Purinergic P2Y Receptor Antagonists/pharmacology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...