Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.014
Filtrar
1.
Function (Oxf) ; 5(3): zqae010, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38706960

RESUMEN

The Olfr78 gene encodes a G-protein-coupled olfactory receptor that is expressed in several ectopic sites. Olfr78 is one of the most abundant mRNA species in carotid body (CB) glomus cells. These cells are the prototypical oxygen (O2) sensitive arterial chemoreceptors, which, in response to lowered O2 tension (hypoxia), activate the respiratory centers to induce hyperventilation. It has been proposed that Olfr78 is a lactate receptor and that glomus cell activation by the increase in blood lactate mediates the hypoxic ventilatory response (HVR). However, this proposal has been challenged by several groups showing that Olfr78 is not a physiologically relevant lactate receptor and that the O2-based regulation of breathing is not affected in constitutive Olfr78 knockout mice. In another study, constitutive Olfr78 knockout mice were reported to have altered systemic and CB responses to mild hypoxia. To further characterize the functional role of Olfr78 in CB glomus cells, we here generated a conditional Olfr78 knockout mouse strain and then restricted the knockout to glomus cells and other catecholaminergic cells by crossing with a tyrosine hydroxylase-specific Cre driver strain (TH-Olfr78 KO mice). We find that TH-Olfr78 KO mice have a normal HVR. Interestingly, glomus cells of TH-Olfr78 KO mice exhibit molecular and electrophysiological alterations as well as a reduced dopamine content in secretory vesicles and neurosecretory activity. These functional characteristics resemble those of CB neuroblasts in wild-type mice. We suggest that, although Olfr78 is not essential for CB O2 sensing, activation of Olfr78-dependent pathways is required for maturation of glomus cells.


Asunto(s)
Cuerpo Carotídeo , Ratones Noqueados , Receptores Odorantes , Tirosina 3-Monooxigenasa , Animales , Cuerpo Carotídeo/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Tirosina 3-Monooxigenasa/genética , Ratones , Hipoxia/metabolismo , Hipoxia/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Masculino , Ratones Endogámicos C57BL
2.
Nihon Yakurigaku Zasshi ; 159(3): 165-168, 2024.
Artículo en Japonés | MEDLINE | ID: mdl-38692881

RESUMEN

Molecular oxygen suffices the ATP production required for the survival of us aerobic organisms. But it is also true that oxygen acts as a source of reactive oxygen species that elicit a spectrum of damages in living organisms. To cope with such intrinsic ambiguity of biological activity oxygen exerts, aerobic mechanisms are equipped with an exquisite adaptive system, which sensitively detects partial pressure of oxygen within the body and controls appropriate oxygen supply to the tissues. Physiological responses to hypoxia are comprised of the acute and chronic phases, in the former of which the oxygen-sensing remains controversial particularly from mechanistic points of view. Recently, we have revealed that the prominently redox-sensitive cation channel TRPA1 plays key roles in oxygen-sensing mechanisms identified in the peripheral tissues and the central nervous system. In this review, we summarize recent development of researches on oxygen-sensing mechanisms including that in the carotid body, which has been recognized as the oxygen receptor organ central to acute oxygen-sensing. We also discuss how ubiquitously the TRPA1 contributes to the mechanisms underlying the acute phase of adaptation to hypoxia.


Asunto(s)
Oxígeno , Canal Catiónico TRPA1 , Canales de Potencial de Receptor Transitorio , Canal Catiónico TRPA1/metabolismo , Humanos , Oxígeno/metabolismo , Animales , Canales de Potencial de Receptor Transitorio/metabolismo , Hipoxia/metabolismo , Canales de Calcio/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Cuerpo Carotídeo/metabolismo
3.
Genes (Basel) ; 15(3)2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38540361

RESUMEN

The carotid body (CB), located bilaterally at the carotid artery bifurcations, is the primary sensory organ for monitoring arterial blood O2 levels. Carotid bodies are immature at birth, exhibiting low sensitivity to hypoxia, and become more sensitive with maturation during the first few weeks of neonatal life. To understand the molecular basis for the postnatal developmental hypoxic responses of CB, we isolated CBs from 5-day and 21-day-old Sprague-Dawley rats and performed RNA sequencing, which allows comprehensive analysis of gene expression. Differentially expressed genes (DEGs) were generated using Edge R, while functional enrichment analysis was performed using gene-set enrichment analysis (GSEA). Analysis of RNA-Seq data showed 2604 DEGs of the total 12,696 genes shared between neonates and adults. Of the 2604 DEGs, 924 genes were upregulated, and 1680 genes were downregulated. Further analysis showed that genes related to oxidative phosphorylation (Ox/phos) and hypoxia-signaling pathways were significantly upregulated in neonatal CBs compared to adult CBs, suggesting a possible link to differential developmental hypoxic responses seen in CB. Genes related to cytokine signaling (INFγ and TNFα) and transcription factors (CREB and NFΚB) mediated pathways were enriched in adult CBs, suggesting that expression of these pathways may be linked to developmental regulation. The RNA-Seq results were verified by analyzing mRNA changes in selected genes by qRT-PCR. Our results of enrichment analysis of biological pathways offer valuable insight into CB hypoxic sensing responses related to the development process.


Asunto(s)
Cuerpo Carotídeo , Ratas , Animales , Cuerpo Carotídeo/metabolismo , Ratas Sprague-Dawley , Perfilación de la Expresión Génica , Hipoxia/metabolismo , Factores de Transcripción/metabolismo
4.
Respir Res ; 25(1): 61, 2024 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-38281036

RESUMEN

BACKGROUND: Peripheral blood oxygen monitoring via chemoreceptors in the carotid body (CB) is an integral function of the autonomic cardiorespiratory regulation. The presence of the purinergic P2Y12 receptor (P2Y12R) has been implicated in CB; however, the exact role of the receptor in O2 sensing and signal transduction is unknown. METHODS: The presence of P2Y12R was established by immunoblotting, RT qPCR and immunohistochemistry. Primary glomus cells were used to assess P2Y12R function during hypoxia and hypercapnia, where monoamines were measured by HPLC; calcium signal was recorded utilizing OGB-1 and N-STORM Super-Resolution System. Ingravescent hypoxia model was tested in anaesthetized mice of mixed gender and cardiorespiratory parameters were recorded in control and receptor-deficient or drug-treated experimental animals. RESULTS: Initially, the expression of P2Y12R in adult murine CB was confirmed. Hypoxia induced a P2Y12R-dependent release of monoamine transmitters from isolated CB cells. Receptor activation with the endogenous ligand ADP promoted release of neurotransmitters under normoxic conditions, while blockade disrupted the amplitude and duration of the intracellular calcium concentration. In anaesthetised mice, blockade of P2Y12R expressed in the CB abrogated the initiation of compensatory cardiorespiratory changes in hypoxic environment, while centrally inhibited receptors (i.e. microglial receptors) or receptor-deficiency induced by platelet depletion had limited influence on the physiological adjustment to hypoxia. CONCLUSIONS: Peripheral P2Y12R inhibition interfere with the complex mechanisms of acute oxygen sensing by influencing the calcium signalling and the release of neurotransmitter molecules to evoke compensatory response to hypoxia. Prospectively, the irreversible blockade of glomic receptors by anti-platelet drugs targeting P2Y12Rs, propose a potential, formerly unrecognized side-effect to anti-platelet medications in patients with pulmonary morbidities.


Asunto(s)
Cuerpo Carotídeo , Humanos , Ratones , Animales , Cuerpo Carotídeo/metabolismo , Oxígeno , Receptores Purinérgicos P2Y12/genética , Receptores Purinérgicos P2Y12/metabolismo , Calcio/metabolismo , Hipoxia/metabolismo
5.
Anat Sci Int ; 99(1): 68-74, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37410337

RESUMEN

In the carotid body of laboratory rodents, adenosine 5'-triphosphate (ATP)-mediated transmission is regarded as critical for transmission from chemoreceptor type I cells to P2X3 purinoceptor-expressing sensory nerve endings. The present study investigated the distribution of P2X3-immunoreactive sensory nerve endings in the carotid body of the adult male Japanese monkey (Macaca fuscata) using multilabeling immunofluorescence. Immunoreactivity for P2X3 was detected in nerve endings associated with chemoreceptor type I cells immunoreactive for synaptophysin. Spherical or flattened terminal parts of P2X3-immunoreactive nerve endings were in close apposition to the perinuclear cytoplasm of synaptophysin-immunoreactive type I cells. Immunoreactivity for ectonucleoside triphosphate diphosphohydrolase 2 (NTPDase2), which hydrolyzes extracellular ATP, was localized in the cell body and cytoplasmic processes of S100B-immunoreactive cells. NTPDase2-immunoreactive cells surrounded P2X3-immunoreactive terminal parts and synaptophysin-immunoreactive type I cells, but did not intrude into attachment surfaces between terminal parts and type I cells. These results suggest ATP-mediated transmission between type I cells and sensory nerve endings in the carotid body of the Japanese monkey, as well as those of rodents.


Asunto(s)
Cuerpo Carotídeo , Ratas , Animales , Masculino , Cuerpo Carotídeo/metabolismo , Macaca fuscata/metabolismo , Receptores Purinérgicos P2X3/metabolismo , Sinaptofisina/metabolismo , Ratas Wistar , Células Receptoras Sensoriales/metabolismo , Adenosina Trifosfato/metabolismo
6.
Purinergic Signal ; 20(1): 5-8, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37211586

RESUMEN

Heart failure is associated with multiple mechanisms, including sympatho-excitation, and is one of the leading causes of death worldwide. Enhanced carotid body chemoreflex function is strongly related to excessive sympathetic nerve activity and sleep-disordered breathing in heart failure. How to reduce the excitability of the carotid body is still scientifically challenging. Both clinical and experimental evidence have suggested that targeting purinergic receptors is of great potential to combat heart failure. In a recent study, Lataro et al. (Lataro et al. in Nat Commun 14:1725, 5) demonstrated that targeting purinergic P2X3 receptors in the carotid body attenuates the progression of heart failure. Using a series of molecular, biochemical, and functional assays, the authors observed that the carotid body generates spontaneous, episodic burst discharges coincident with the onset of disordered breathing in male rats with heart failure, which was generated by ligating the left anterior descending coronary artery. Moreover, P2X3 receptor expression was found to be upregulated in the petrosal ganglion chemoreceptive neurons of rats with heart failure. Of particular note, treatment with a P2X3 antagonist rescued pathological breathing disturbances, abolished episodic discharges, reinstated autonomic balance, attenuated cardiac dysfunction, and reduced the immune cell response and plasma cytokine levels in those rats.


Asunto(s)
Cuerpo Carotídeo , Insuficiencia Cardíaca , Ratas , Masculino , Animales , Cuerpo Carotídeo/metabolismo , Receptores Purinérgicos P2X/metabolismo , Insuficiencia Cardíaca/metabolismo , Neuronas/metabolismo , Sistema Nervioso Simpático , Receptores Purinérgicos P2X3/metabolismo , Receptores Purinérgicos P2X2/metabolismo
7.
Exp Physiol ; 109(4): 461-469, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38031809

RESUMEN

An adequate supply of O2 is essential for the maintenance of cellular activity. Systemic or local hypoxia can be experienced during decreased O2 availability or associated with diseases, or a combination of both. Exposure to hypoxia triggers adjustments in multiple physiological systems in the body to generate appropriate homeostatic responses. However, with significant reductions in the arterial partial pressure of O2, hypoxia can be life-threatening and cause maladaptive changes or cell damage and death. To mitigate the impact of limited O2 availability on cellular activity, O2 chemoreceptors rapidly detect and respond to reductions in the arterial partial pressure of O2, triggering orchestrated responses of increased ventilation and cardiac output, blood flow redistribution and metabolic adjustments. In mammals, the peripheral chemoreceptors of the carotid body are considered to be the main hypoxic sensors and the primary source of excitatory feedback driving respiratory, cardiovascular and autonomic responses. However, current evidence indicates that the CNS contains specialized brainstem and spinal cord regions that can also sense hypoxia and stimulate brain networks independently of the carotid body inputs. In this manuscript, we review the discoveries about the functioning of the O2 chemoreceptors and their contribution to the monitoring of O2 levels in the blood and brain parenchyma and mounting cardiorespiratory responses to maintain O2 homeostasis. We also discuss the implications of the chemoreflex-related mechanisms in paediatric and adult pathologies.


Asunto(s)
Cuerpo Carotídeo , Hipoxia , Animales , Humanos , Niño , Células Quimiorreceptoras/fisiología , Cuerpo Carotídeo/metabolismo , Respiración , Pulmón , Mamíferos/metabolismo , Oxígeno/metabolismo
8.
J Appl Physiol (1985) ; 136(2): 233-243, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38126089

RESUMEN

The carotid bodies (CBs) have been implicated in glucose abnormalities in obesity via elevation of activity of the sympathetic nervous system. Obesity-induced hypertension is mediated by insulin receptor (INSR) signaling and by leptin, which binds to the leptin receptor (LEPRb) in CB and activates transient receptor potential channel subfamily M member 7 (TRPM7). We hypothesize that in mice with diet-induced obesity, hyperglycemia, glucose intolerance, and insulin resistance will be attenuated by the CB denervation (carotid sinus nerve dissection, CSND) and by knockdown of Leprb, Trpm7, and Insr gene expression in CB. In series of experiments in 75 male diet-induced obese (DIO) mice, we performed either CSND (vs. sham) surgeries or shRNA-induced suppression of Leprb, Trpm7, or Insr gene expression in CB, followed by blood pressure telemetry, intraperitoneal glucose tolerance and insulin tolerance tests, and measurements of fasting plasma insulin, leptin, corticosterone, glucagon and free fatty acids (FFAs) levels, hepatic expression of gluconeogenesis enzymes phosphoenolpyruvate carboxykinase (PEPCK) and glucose 6-phosphatase (G-6-Pase) mRNA and liver glycogen levels. CSND decreased blood pressure, fasting blood glucose levels and improved glucose tolerance without any effect on insulin resistance. CSND did not affect any hormone levels and gluconeogenesis enzymes, but increased liver glycogen level. Genetic knockdown of CB Leprb, Trpm7, and Insr had no effect on glucose metabolism. We conclude that CB contributes to hyperglycemia of obesity, probably by modulation of the glycogen-glucose equilibrium. Diabetogenic effects of obesity on CB in mice do not occur via activation of CB Leprb, Trpm7, and Insr.NEW & NOTEWORTHY This paper provides first evidence that carotid body denervation abolishes hypertension and improves fasting blood glucose levels and glucose tolerance in mice with diet-induced obesity. Furthermore, we have shown that this phenomenon is associated with increased liver glycogen content, whereas insulin sensitivity and enzymes of gluconeogenesis were not affected.


Asunto(s)
Cuerpo Carotídeo , Hiperglucemia , Hipertensión , Resistencia a la Insulina , Insulinas , Canales Catiónicos TRPM , Masculino , Ratones , Animales , Leptina , Glucemia/metabolismo , Cuerpo Carotídeo/metabolismo , Ratones Obesos , Canales Catiónicos TRPM/metabolismo , Glucógeno Hepático/metabolismo , Hiperglucemia/metabolismo , Obesidad/metabolismo , Glucosa/metabolismo , Hipertensión/metabolismo , Desnervación , Insulinas/metabolismo
9.
Int J Mol Sci ; 24(24)2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-38138991

RESUMEN

The carotid body is a major peripheral chemoreceptor that senses changes in arterial blood oxygen, carbon dioxide, and pH, which is important for the regulation of breathing and cardiovascular function. The mechanisms by which the carotid body senses O2 and CO2 are well known; conversely, the mechanisms by which it senses pH variations are almost unknown. Here, we used immunohistochemistry to investigate how the human carotid body contributes to the detection of acidosis, analyzing whether it expresses acid-sensing ion channels (ASICs) and determining whether these channels are in the chemosensory glomic cells or in the afferent nerves. In ASIC1, ASIC2, and ASIC3, and to a much lesser extent ASIC4, immunoreactivity was detected in subpopulations of type I glomus cells, as well as in the nerves of the carotid body. In addition, immunoreactivity was found for all ASIC subunits in the neurons of the petrosal and superior cervical sympathetic ganglia, where afferent and efferent neurons are located, respectively, innervating the carotid body. This study reports for the first time the occurrence of ASIC proteins in the human carotid body, demonstrating that they are present in glomus chemosensory cells (ASIC1 < ASIC2 > ASIC3 > ASIC4) and nerves, presumably in both the afferent and efferent neurons supplying the organ. These results suggest that the detection of acidosis by the carotid body can be mediated via the ASIC ion channels present in the type I glomus cells or directly via sensory nerve fibers.


Asunto(s)
Acidosis , Cuerpo Carotídeo , Humanos , Canales Iónicos Sensibles al Ácido/metabolismo , Cuerpo Carotídeo/metabolismo , Células Quimiorreceptoras/metabolismo , Sistema Nervioso Periférico/metabolismo , Acidosis/metabolismo
10.
Adv Anat Embryol Cell Biol ; 237: 13-35, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37946075

RESUMEN

The carotid body (CB) is the main peripheral arterial chemoreceptor that registers the levels of pO2, pCO2 and pH in the blood and responds to their changes by regulating breathing. It is strategically located in the bifurcation of each common carotid artery. The organ consists of "glomera" composed of two cell types, glomus and sustentacular cells, interspersed by blood vessels and nerve bundles and separated by connective tissue. The neuron-like glomus or type I cells are considered as the chemosensory cells of the CB. They contain numerous cytoplasmic organelles and dense-cored vesicles that store and release neurotransmitters. They also form both conventional chemical and electrical synapses between each other and are contacted by peripheral nerve endings of petrosal ganglion neurons. The glomus cells are dually innervated by both sensory nerve fibers through the carotid sinus nerve and autonomic fibers of sympathetic origin via the ganglioglomerular nerve. The parasympathetic efferent innervation is relayed by vasomotor fibers of ganglion cells located around or inside the CB. The glial-like sustentacular or type II cells are regarded to be supporting cells although they sustain physiologic neurogenesis in the adult CB and are thus supposed to be progenitor cells as well. The CB is a highly vascularized organ and its intraorgan hemodynamics possibly plays a role in the process of chemoreception.


Asunto(s)
Cuerpo Carotídeo , Animales , Cuerpo Carotídeo/metabolismo , Células Quimiorreceptoras/fisiología , Neuronas , Arteria Carótida Común , Ganglios , Mamíferos
11.
Adv Anat Embryol Cell Biol ; 237: 37-48, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37946076

RESUMEN

The mammalian carotid body (CB) exhibits considerable plasticity of its structure during development and aging and as a consequence of environmental, metabolic and inflammatory stimuli. The structural changes during maturation include an enlargement of the total and vascular volume of the CB. Conversely, aging results in a reduction in the number and volume of glomus cells with progressive cellular degeneration and an apparent increase in the surrounding connective tissue. Age-related structural alterations are similar to those during chronic hypoxia. Long-term hypoxic exposure and sodium nitrate treatment enlarge several-fold the size of the rat CB causing glomus cell hypertrophy and hyperplasia, and evoke changes in its vascular structure, inducing marked vasodilation and neovascularization. In humans, such structural CB adaptation responses to prolonged hypoxia occur during acclimatization to high altitudes. On the other hand, the hyperoxic CB is significantly smaller than those of age-matched normoxic controls. Morphological alterations in the CB in both hypertensive animals and humans are characterized by a slightly enlarged parenchyma without apparent vascular expansion and/or dilation. The CB structural plasticity depends on the existence of a population of multipotent neural crest-derived stem cells, which are activated during hypoxia to proliferate and differentiate into new both neuronal (glomus) and vascular cell types.


Asunto(s)
Cuerpo Carotídeo , Humanos , Ratas , Animales , Cuerpo Carotídeo/metabolismo , Hipoxia/metabolismo , Neuronas/metabolismo , Neovascularización Patológica/metabolismo , Mamíferos
12.
Adv Anat Embryol Cell Biol ; 237: 105-122, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37946079

RESUMEN

A striking feature of the carotid body (CB) is its remarkable degree of plasticity in a variety of neurotransmitter/modulator systems in response to environmental stimuli, particularly following hypoxic exposure of animals and during ascent to high altitude. Current evidence suggests that acetylcholine and adenosine triphosphate are two major excitatory neurotransmitter candidates in the hypoxic CB, and they may also be involved as co-transmitters in hypoxic signaling. Conversely, dopamine, histamine and nitric oxide have recently been considered inhibitory transmitters/modulators of hypoxic chemosensitivity. It has also been revealed that interactions between excitatory and inhibitory messenger molecules occur during hypoxia. On the other hand, alterations in purinergic neurotransmitter mechanisms have been implicated in ventilatory acclimatization to hypoxia. Chronic hypoxia also induces profound changes in other neurochemical systems within the CB such as the catecholaminergic, peptidergic and nitrergic, which in turn may contribute to increased ventilatory and chemoreceptor responsiveness to hypoxia at high altitude. Taken together, current data suggest that complex interactions among transmitters markedly influence hypoxia-induced transmitter release from the CB. In addition, the expression of a wide variety of growth factors, proinflammatory cytokines and their receptors have been identified in CB parenchymal cells in response to hypoxia and their upregulated expression could mediate the local inflammation and functional alteration of the CB under hypoxic conditions.


Asunto(s)
Cuerpo Carotídeo , Animales , Cuerpo Carotídeo/metabolismo , Células Quimiorreceptoras/metabolismo , Hipoxia/metabolismo , Adenosina Trifosfato/metabolismo , Neurotransmisores/metabolismo
13.
Adv Anat Embryol Cell Biol ; 237: 139-153, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37946081

RESUMEN

Accumulating evidence suggests that the mammalian carotid body (CB) constitutes a neurogenic center that contains a functionally active germinal niche. A variety of transcription factors is required for the generation of a precursor cell pool in the developing CB. Most of them are later silenced in their progeny, thus allowing for the maturation of the differentiated neurons. In the adult CB, neurotransmitters and vascular cytokines released by glomus cells upon exposure to chronic hypoxia act as paracrine signals that induce proliferation and differentiation of pluripotent stem cells, neuronal and vascular progenitors. Key proliferation markers such as Ki-67 and BrdU are widely used to evaluate the proliferative status of the CB parenchymal cells in the initial phase of this neurogenesis. During hypoxia sustentacular cells which are dormant cells in normoxic conditions can proliferate and differentiate into new glomus cells. However, more recent data have revealed that the majority of the newly formed glomus cells is derived from the glomus cell lineage itself. The mature glomus cells express numerous trophic and growth factors, and their corresponding receptors, which act on CB cell populations in autocrine or paracrine ways. Some of them initially serve as target-derived survival factors and then as signaling molecules in developing vascular targets. Morphofunctional insights into the cellular interactions in the CB stem cell microenvironment can be helpful in further understanding the therapeutic potential of the CB cell niche.


Asunto(s)
Cuerpo Carotídeo , Nicho de Células Madre , Animales , Cuerpo Carotídeo/metabolismo , Neuronas/metabolismo , Diferenciación Celular , Hipoxia/metabolismo , Mamíferos
14.
Adv Anat Embryol Cell Biol ; 237: 63-103, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37946078

RESUMEN

Carotid body (CB) glomus cells in most mammals, including humans, contain a broad diversity of classical neurotransmitters, neuropeptides and gaseous signaling molecules as well as their cognate receptors. Among them, acetylcholine, adenosine triphosphate and dopamine have been proposed to be the main excitatory transmitters in the mammalian CB, although subsequently dopamine has been considered an inhibitory neuromodulator in almost all mammalian species except the rabbit. In addition, co-existence of biogenic amines and neuropeptides has been reported in the glomus cells, thus suggesting that they store and release more than one transmitter in response to natural stimuli. Furthermore, certain metabolic and transmitter-degrading enzymes are involved in the chemotransduction and chemotransmission in various mammals. However, the presence of the corresponding biosynthetic enzyme for some transmitter candidates has not been confirmed, and neuroactive substances like serotonin, gamma-aminobutyric acid and adenosine, neuropeptides including opioids, substance P and endothelin, and gaseous molecules such as nitric oxide have been shown to modulate the chemosensory process through direct actions on glomus cells and/or by producing tonic effects on CB blood vessels. It is likely that the fine balance between excitatory and inhibitory transmitters and their complex interactions might play a more important than suggested role in CB plasticity.


Asunto(s)
Cuerpo Carotídeo , Neuropéptidos , Humanos , Animales , Conejos , Cuerpo Carotídeo/metabolismo , Dopamina/metabolismo , Neurotransmisores/metabolismo , Neuropéptidos/metabolismo , Mamíferos
15.
Adv Anat Embryol Cell Biol ; 237: 155-159, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37946082

RESUMEN

During the past decade, the carotid body (CB) has been considered an innovative therapeutic target for the treatment of certain cardiorespiratory and metabolic diseases most of which are sympathetically mediated. It has recently been revealed that CB stem cells provide new target sites for the development of promising cell-based therapies. Specifically, generation of CB progenitors in vitro which can differentiate into functionally active glomus cells may be a useful procedure to produce the cell mass required for replacement cell therapy. Due to their dopaminergic nature, adult glomus cells can be used for an intrastriatal grafting in neurodegenerative brain disorders including Parkinson's disease. The beneficial effect of throphic factors such as glial cell-derived neurotrophic factor synergistically released by the transplanted cells then enables the transplant to survive. Likewise, intracerebral administration of CB cell aggregates or dispersed cells has been tested for the treatment of an experimental model of stroke. The systematic clinical applicability of CB autotransplants following glomectomy in humans is under investigation. In such autotransplantation studies, cell aggregates from unilaterally resected CB might be used as autografts. In addition, stem cells could offer an opportunity for tissue expansion and might settle the issue of small number of glomus cells available for transplantation.


Asunto(s)
Cuerpo Carotídeo , Enfermedad de Parkinson , Adulto , Humanos , Cuerpo Carotídeo/metabolismo , Cuerpo Carotídeo/trasplante , Enfermedad de Parkinson/metabolismo , Neuronas/metabolismo , Dopamina/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos
16.
Sheng Li Xue Bao ; 75(5): 629-635, 2023 Oct 25.
Artículo en Chino | MEDLINE | ID: mdl-37909134

RESUMEN

The purpose of the present study was to explore the role of carotid body metabotropic glutamate receptor 1 (mGluR1) in chronic intermittent hypoxia (CIH)-induced carotid body plasticity. Sprague Dawley (SD) rats were exposed to CIH (6%-21% O2, 4 min/cycle, 8 h/day) for 4 weeks. The blood pressure of rats was monitored non-invasively by tail-cuff method under consciousness. RT-qPCR was used to examine the mRNA expression level of mGluR1 in rat carotid body. Western blot was used to detect the protein expression level of mGluR1 in rat carotid body. The role of mGluR1 in CIH-induced carotid body sensory long-term facilitation (sLTF) was investigated by ex vivo carotid sinus nerve discharge recording, and the carotid body sLTF was evoked by a 10-episode of repetitive acute intermittent hypoxia (AIH: 1 min of 5% O2 interspersed with 5 min of 95% O2). The results showed that: 1) CIH increased the systolic blood pressure (P < 0.001), diastolic blood pressure (P < 0.005) and mean arterial blood pressure (P < 0.001) of rats; 2) CIH decreased the mRNA and protein levels of mGluR1 in the rat carotid body (P < 0.01); 3) 4 weeks of CIH induced carotid body sLTF significantly, exhibiting as an increasing baseline sensory activity during post-AIH, which was inhibited by application of an agonist of group I metabotropic glutamate receptors, (S)-3,5-dihydroxyphenylglycine (DHPG), during sLTF induction (P < 0.005). In summary, these results suggest that activation of mGluR1 inhibits CIH-induced carotid body plasticity in rats.


Asunto(s)
Cuerpo Carotídeo , Receptores de Glutamato Metabotrópico , Ratas , Animales , Cuerpo Carotídeo/metabolismo , Ratas Sprague-Dawley , Hipoxia , Receptores de Glutamato Metabotrópico/metabolismo , ARN Mensajero/metabolismo
17.
Sheng Li Xue Bao ; 75(4): 529-536, 2023 Aug 25.
Artículo en Chino | MEDLINE | ID: mdl-37583040

RESUMEN

The aim of the present study was to explore the role of group II and III metabotropic glutamate receptors (mGluRs) in carotid body plasticity induced by chronic intermittent hypoxia (CIH) in rats. Sprague Dawley (SD) rats were treated with CIH in Oxycycler A84 hypoxic chamber for 4 weeks, and the tail artery blood pressure was measured at the end of model preparation. RT-qPCR was performed to examine the mRNA expression levels of mGluR2/3/8 in rat carotid body. Carotid sinus nerve activity was detected by ex vivo carotid sinus nerve discharge recording technique, and acute intermittent hypoxia (AIH) was administered to induce carotid body sensory long-term facilitation (sLTF), in order to observe the role of group II and group III mGluRs in carotid body plasticity induced by CIH. The results showed that: 1) After 4 weeks of CIH exposure, the blood pressure of rats increased significantly; 2) CIH down-regulated the mRNA levels of mGluR2/3, and up-regulated the mRNA level of mGluR8 in the carotid body; 3) AIH induced sLTF in carotid body of CIH group. In the CIH group, activation of group II mGluRs had no effect on sLTF of carotid body, while activation of group III mGluRs completely inhibited sLTF. These results suggest that CIH increases blood pressure in rats, and group III mGluRs play an inhibitory role in CIH-induced carotid body plasticity in rats.


Asunto(s)
Cuerpo Carotídeo , Receptores de Glutamato Metabotrópico , Ratas , Animales , Cuerpo Carotídeo/metabolismo , Ratas Sprague-Dawley , Hipoxia , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , ARN Mensajero/metabolismo
18.
Sci Adv ; 9(27): eadf3026, 2023 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-37406126

RESUMEN

Oxygen (O2) sensing by the carotid body is critical for maintaining cardiorespiratory homeostasis during hypoxia. Hydrogen sulfide (H2S) signaling is implicated in carotid body activation by low O2. Here, we show that persulfidation of olfactory receptor 78 (Olfr78) by H2S is an integral component of carotid body activation by hypoxia. Hypoxia and H2S increased persulfidation in carotid body glomus cells and persulfidated cysteine240 in Olfr78 protein in heterologous system. Olfr78 mutants manifest impaired carotid body sensory nerve, glomus cell, and breathing responses to H2S and hypoxia. Glomus cells are positive for GOlf, adenylate cyclase 3 (Adcy3) and cyclic nucleotide-gated channel alpha 2 (Cnga2), key molecules of odorant receptor signaling. Adcy3 or Cnga2 mutants exhibited impaired carotid body and glomus cell responses to H2S and breathing responses to hypoxia. These results suggest that H2S through redox modification of Olfr78 participates in carotid body activation by hypoxia to regulate breathing.


Asunto(s)
Cuerpo Carotídeo , Sulfuro de Hidrógeno , Receptores Odorantes , Humanos , Receptores Odorantes/metabolismo , Hipoxia/metabolismo , Sulfuro de Hidrógeno/metabolismo , Cuerpo Carotídeo/metabolismo , Oxígeno/metabolismo
19.
Acta Histochem ; 125(4): 152043, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37126880

RESUMEN

Carotid body (CB) activity stimulated by a lower partial oxygen pressure in rats is enhanced by exposure to chronic intermittent hypoxia. However, the mechanisms that modulate CB activity remain unclear. In the present study, the expression and distribution of one of the candidate molecules to modulate reactivity, Ca2+/calmodulin-dependent protein kinase II (CaMKII) were examined in the rat CB using reverse transcriptional polymerase chain reaction and immunofluorescence with isoform-specific antibodies. CaMKIIγ and CaMKIIδ were distributed in CB chemoreceptor cells, and exhibited intense immunoreactivity in dopamine ß-hydroxylase-positive chemoreceptor cells. CaMKIIß and CaMKIIγ were distributed in sensory nerve endings attached to chemoreceptor cells of the CB. In the petrosal ganglion, immunoreactivities for CaMKIIα, CaMKIIß, CaMKIIγ, and CaMKIIδ were detected in the perinuclear region of ganglion cells. The present results indicate that CaMKIIγ and CaMKIIδ in chemoreceptor cells and CaMKIIß and CaMKIIγ in sensory nerve endings enhanced reciprocal synaptic transmission, i.e., noradrenaline and ATP for cells to neurons and glutamate for neurons to cells.


Asunto(s)
Cuerpo Carotídeo , Ratas , Animales , Cuerpo Carotídeo/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Células Quimiorreceptoras , Neuronas/metabolismo , Transmisión Sináptica/fisiología
20.
Respir Physiol Neurobiol ; 313: 104063, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37076025

RESUMEN

The carotid body (CB) is a neuroepithelial tissue consisting of O2-sensitive glomus cells that constantly scan the arterial blood for O2 and generate a discharge as an inverse function of O2 content. Aging is a cumulative result of decreased O2 supply paralleled by a decreased O2 tissue demand and oxidative damage to cells derived from aerobic metabolism. Here we studied how CB affects the aging process. This is a study of CB ultrastructural morphometry and immunohistochemical expression of proteins underlying CB responsiveness. The study was based on human CBs obtained from cadavers of people who died due to traumatic events in young and old age. The study was supplemented by investigations of CBs obtained from young and old rats subjected to chronic normoxic and hypoxic conditions. We found changes in the old normoxic CBs akin to the effects of chronic hypoxia such as enhanced extracellular matrix, reduced synaptic contacts between glomus cells, fewer glomus cells, secretory vesicles, and mitochondria. These changes were accompanied by enhanced expressions of hypoxia-inducible factor one-alpha (HIF-1α), vascular endothelial growth factor (VEGF), and nitric oxide synthase (NOS2). We conclude that hypoxia and aging share a common background consisting of deficient O2 tissue supply, mitochondrial dysfunction, and a limited ability to deal with increased cellular oxidative stress. Aging leads to adaptative reductions in CB responsiveness to hypoxia shifting the chemosensory setpoint upward. We submit that the attenuated CB sensitivity at old age may be tantamount to "physiological denervation" leading to a gradual loss of the chemosensing role in the prevention of tissue hypoxia by increasing lung ventilation.


Asunto(s)
Cuerpo Carotídeo , Ratas , Humanos , Animales , Cuerpo Carotídeo/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Hipoxia , Óxido Nítrico Sintasa/metabolismo , Envejecimiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...