Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Physiol ; 596(15): 2969-2976, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29214644

RESUMO

The molecular underpinnings of the oxygen sensitivity of the carotid body Type I cells are becoming better defined as research begins to identify potential interactions between previously separate theories. Nevertheless, the field of oxygen chemoreception still presents the general observer with a bewildering array of potential signalling pathways by which a fall in oxygen levels might initiate Type I cell activation. The purpose of this brief review is to address five of the current oxygen sensing hypotheses: the lactate-Olfr 78 hypothesis of oxygen chemotransduction; the role mitochondrial ATP and metabolism may have in chemotransduction; the AMP-activated protein kinase hypothesis and its current role in oxygen sensing by the carotid body; reactive oxygen species as key transducers in the oxygen sensing cascade; and the mechanisms by which H2 S, reactive oxygen species and haem oxygenase may integrate to provide a rapid oxygen sensing transduction system. Over the previous 15 years several lines of research into acute hypoxic chemotransduction mechanisms have focused on the integration of mitochondrial and membrane signalling. This review places an emphasis on the subplasmalemmal-mitochondrial microenvironment in Type I cells and how theories of acute oxygen sensing are increasingly dependent on functional interaction within this microenvironment.


Assuntos
Corpo Carotídeo/fisiologia , Oxigênio/fisiologia , Animais
2.
Adv Exp Med Biol ; 1071: 137-142, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30357744

RESUMO

The carotid bodies (CB) respond to changes in blood gases with neurotransmitter release, thereby increasing carotid sinus nerve firing frequency and ultimately correcting the pattern of breathing. It has previously been demonstrated that acute application of the adipokine leptin augments the hypoxic sensory response of the intact in-vitro CB (Pye RL, Roy A, Wilson RJ, Wyatt CN. FASEB J 30(1 Supplement):983.1, 2016) and isolated CB type I cell (Pye RL, Dunn EJ, Ricker EM, Jurcsisn JG, Barr BL, Wyatt CN. Arterial chemoreceptors in physiology and pathophysiology. Advances in experimental medicine and biology. Springer, Cham, 2015). This study's aim was to examine, in-vivo, if elevated leptin modulated CB function and breathing.Rats were fed high fat or control chow for 16-weeks. High fat fed (HFF) animals gained significantly more weight compared to control fed (CF) animals and had significantly higher serum leptin levels compared to CF. Utilizing whole-body plethysmography, HFF animals demonstrated significantly depressed breathing compared to CF at rest and during hypoxia. However, amplitudes in the change in breathing from rest to hypoxia were not significantly different between groups. CB type I cells were isolated and intracellular calcium levels recorded. Averaged and peak cellular hypoxic responses were not significantly different.Despite a small but significant rise in leptin, differences in breathing caused by high fat feeding are unlikely caused by an effect of leptin on CB type I cells. However, the possibility remains that leptin may have in-vivo postsynaptic effects on the carotid sinus nerve; this remains to be investigated.


Assuntos
Corpo Carotídeo/fisiopatologia , Células Quimiorreceptoras/citologia , Dieta Hiperlipídica , Hipóxia/fisiopatologia , Respiração , Animais , Gasometria , Ratos
3.
J Physiol ; 594(15): 4439-52, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27027261

RESUMO

KEY POINTS: Hypoglycaemia is counteracted by release of hormones and an increase in ventilation and CO2 sensitivity to restore blood glucose levels and prevent a fall in blood pH. The full counter-regulatory response and an appropriate increase in ventilation is dependent on carotid body stimulation. We show that the hypoglycaemia-induced increase in ventilation and CO2 sensitivity is abolished by preventing adrenaline release or blocking its receptors. Physiological levels of adrenaline mimicked the effect of hypoglycaemia on ventilation and CO2 sensitivity. These results suggest that adrenaline, rather than low glucose, is an adequate stimulus for the carotid body-mediated changes in ventilation and CO2 sensitivity during hypoglycaemia to prevent a serious acidosis in poorly controlled diabetes. ABSTRACT: Hypoglycaemia in vivo induces a counter-regulatory response that involves the release of hormones to restore blood glucose levels. Concomitantly, hypoglycaemia evokes a carotid body-mediated hyperpnoea that maintains arterial CO2 levels and prevents respiratory acidosis in the face of increased metabolism. It is unclear whether the carotid body is directly stimulated by low glucose or by a counter-regulatory hormone such as adrenaline. Minute ventilation was recorded during infusion of insulin-induced hypoglycaemia (8-17 mIU kg(-1)  min(-1) ) in Alfaxan-anaesthetised male Wistar rats. Hypoglycaemia significantly augmented minute ventilation (123 ± 4 to 143 ± 7 ml min(-1) ) and CO2 sensitivity (3.3 ± 0.3 to 4.4 ± 0.4 ml min(-1)  mmHg(-1) ). These effects were abolished by either ß-adrenoreceptor blockade with propranolol or adrenalectomy. In this hypermetabolic, hypoglycaemic state, propranolol stimulated a rise in P aC O2, suggestive of a ventilation-metabolism mismatch. Infusion of adrenaline (1 µg kg(-1)  min(-1) ) increased minute ventilation (145 ± 4 to 173 ± 5 ml min(-1) ) without altering P aC O2 or pH and enhanced ventilatory CO2 sensitivity (3.4 ± 0.4 to 5.1 ± 0.8 ml min(-1)  mmHg(-1) ). These effects were attenuated by either resection of the carotid sinus nerve or propranolol. Physiological concentrations of adrenaline increased the CO2 sensitivity of freshly dissociated carotid body type I cells in vitro. These findings suggest that adrenaline release can account for the ventilatory hyperpnoea observed during hypoglycaemia by an augmented carotid body and whole body ventilatory CO2 sensitivity.


Assuntos
Dióxido de Carbono/fisiologia , Corpo Carotídeo/fisiologia , Epinefrina/fisiologia , Hipoglicemia/fisiopatologia , Ventilação Pulmonar/fisiologia , Antagonistas Adrenérgicos beta/farmacologia , Animais , Cálcio/fisiologia , Hiperinsulinismo/fisiopatologia , Masculino , Propranolol/farmacologia , Ratos Wistar
4.
Nature ; 459(7246): 596-600, 2009 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-19387438

RESUMO

Ca(2+) mobilization from intracellular stores represents an important cell signalling process that is regulated, in mammalian cells, by inositol-1,4,5-trisphosphate (InsP(3)), cyclic ADP ribose and nicotinic acid adenine dinucleotide phosphate (NAADP). InsP(3) and cyclic ADP ribose cause the release of Ca(2+) from sarcoplasmic/endoplasmic reticulum stores by the activation of InsP(3) and ryanodine receptors (InsP(3)Rs and RyRs). In contrast, the nature of the intracellular stores targeted by NAADP and the molecular identity of the NAADP receptors remain controversial, although evidence indicates that NAADP mobilizes Ca(2+) from lysosome-related acidic compartments. Here we show that two-pore channels (TPCs) comprise a family of NAADP receptors, with human TPC1 (also known as TPCN1) and chicken TPC3 (TPCN3) being expressed on endosomal membranes, and human TPC2 (TPCN2) on lysosomal membranes when expressed in HEK293 cells. Membranes enriched with TPC2 show high affinity NAADP binding, and TPC2 underpins NAADP-induced Ca(2+) release from lysosome-related stores that is subsequently amplified by Ca(2+)-induced Ca(2+) release by InsP(3)Rs. Responses to NAADP were abolished by disrupting the lysosomal proton gradient and by ablating TPC2 expression, but were only attenuated by depleting endoplasmic reticulum Ca(2+) stores or by blocking InsP(3)Rs. Thus, TPCs form NAADP receptors that release Ca(2+) from acidic organelles, which can trigger further Ca(2+) signals via sarcoplasmic/endoplasmic reticulum. TPCs therefore provide new insights into the regulation and organization of Ca(2+) signals in animal cells, and will advance our understanding of the physiological role of NAADP.


Assuntos
Canais de Cálcio/metabolismo , Sinalização do Cálcio , Cálcio/metabolismo , NADP/análogos & derivados , Organelas/metabolismo , Animais , Canais de Cálcio/genética , Sinalização do Cálcio/efeitos dos fármacos , Linhagem Celular , Galinhas , Humanos , Concentração de Íons de Hidrogênio , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , NADP/metabolismo , NADP/farmacologia , Organelas/efeitos dos fármacos , Ligação Proteica
5.
Adv Exp Med Biol ; 860: 49-54, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26303466

RESUMO

It is known that opioids inhibit the hypoxic ventilatory response in part via an action at the carotid body, but little is known about the cellular mechanisms that underpin this. This study's objectives were to examine which opioid receptors are located on the oxygen-sensing carotid body type I cells from the rat and determine the mechanism by which opioids might inhibit cellular excitability.Immunocytochemistry revealed the presence of µ and κ opioid receptors on type I cells. The µ-selective agonist DAMGO (10 µM) and the κ-selective agonist U50-488 (10 µM) inhibited high K(+) induced rises in intracellular Ca(2+) compared with controls. After 3 h incubation (37 °C) with pertussis toxin (150 ng ml(-1)), DAMGO (10 µM) and U50-488 (10 µM) had no significant effect on the Ca(2+) response to high K(+).These results indicate that opioids acting at µ and κ receptors inhibit voltage-gated Ca(2+) influx in rat carotid body type I cells via G(i)-coupled mechanisms. This mechanism may contribute to opioid's inhibitory actions in the carotid body.


Assuntos
Cálcio/metabolismo , Corpo Carotídeo/metabolismo , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Animais , Animais Recém-Nascidos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Ratos , Ratos Sprague-Dawley
6.
Adv Exp Med Biol ; 860: 17-24, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26303463

RESUMO

It has previously been reported that AMP-activated protein kinase (AMPK) may be critical for hypoxic chemotransduction in carotid body type I cells. This study sought to determine the importance of the regulatory upstream kinase of AMPK, CamKKß, in the acute response to hypoxia in isolated mouse type I cells.Initial data indicated several previously unreported artefacts associated with using the CamKKß inhibitor STO609 and Ca(2+) imaging techniques. Most importantly Fura-2 and X-Rhod1 imaging revealed that STO609 quenched emission fluorescence even in the absence of intracellular Ca(2+) ([Ca(2+)](I)). Furthermore, STO609 (100 µM) rapidly inhibited outward macroscopic currents and this inhibition was abolished in the presence of the selective BK(Ca) inhibitor paxilline.Taken together these data suggest that ST0609 should be used with caution during Ca(2+) imaging studies as it can directly interact with Ca(2+) binding dyes. The rapid inhibitory effect of STO609 on BK(Ca) was unexpected as the majority of studies using this compound required an incubation of approximately 10 min to inhibit the kinase. Furthermore, as AMPK activation inhibits BK(Ca), inhibiting AMPK's upstream kinases would, if anything, be predicted to have the opposite effect on BK(Ca). Future work will determine if the inhibition of BK(Ca) is via CamKKß or via an off target action of STO609 on the channel itself.


Assuntos
Benzimidazóis/farmacologia , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Cálcio/metabolismo , Corpo Carotídeo/efeitos dos fármacos , Naftalimidas/farmacologia , Canais de Potássio Cálcio-Ativados/antagonistas & inibidores , Animais , Artefatos , Corpo Carotídeo/metabolismo , Linhagem Celular Tumoral , Camundongos
7.
Adv Exp Med Biol ; 860: 61-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26303468

RESUMO

Obesity related pathologies are the health care crisis of our generation. The fat cell derived adipokine leptin has been shown to be a central stimulant of respiration. Very high levels of leptin, however, are associated with the depressed ventilatory phenotype observed in obesity hypoventilation syndrome. Leptin receptors have been identified on carotid body type I cells but how their activation might influence the physiology of these cells is not known.The acute application of leptin evoked calcium signaling responses in isolated type I cells. Cells increased their Fura 2 ratio by 0.074 ± 0.010 ratio units (n = 39, P < 0.001). Leptin also increased the peak membrane currents in 6 of 9 cells increasing the peak macroscopic currents at +10 mV by 61 ± 14 % (p < 0.02). Leptin administered in the presence of the selective BK(Ca) channel inhibitor Paxilline (0.5 µM) failed to increase membrane currents (n = 5). Interestingly, leptin did not significantly alter the resting membrane potential of isolated type I cells (n = 9) and anoxic/acidic depolarizations were unaffected by leptin (n = 7, n = 6).These data suggest that leptin receptors are functional in type I cells but that their acute activation does not alter chemosensory properties. Future studies will use chronic models of leptin dysregulation.


Assuntos
Cálcio/metabolismo , Corpo Carotídeo/efeitos dos fármacos , Leptina/farmacologia , Canais de Potássio Cálcio-Ativados/efeitos dos fármacos , Animais , Corpo Carotídeo/citologia , Corpo Carotídeo/fisiologia , Canais de Potássio Cálcio-Ativados/fisiologia , Ratos , Ratos Sprague-Dawley
8.
J Biol Chem ; 286(14): 11929-36, 2011 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-21209098

RESUMO

Inhibition of large conductance calcium-activated potassium (BKCa) channels mediates, in part, oxygen sensing by carotid body type I cells. However, BKCa channels remain active in cells that do not serve to monitor oxygen supply. Using a novel, bacterially derived AMP-activated protein kinase (AMPK), we show that AMPK phosphorylates and inhibits BKCa channels in a splice variant-specific manner. Inclusion of the stress-regulated exon within BKCa channel α subunits increased the stoichiometry of phosphorylation by AMPK when compared with channels lacking this exon. Surprisingly, however, the increased phosphorylation conferred by the stress-regulated exon abolished BKCa channel inhibition by AMPK. Point mutation of a single serine (Ser-657) within this exon reduced channel phosphorylation and restored channel inhibition by AMPK. Significantly, RT-PCR showed that rat carotid body type I cells express only the variant of BKCa that lacks the stress-regulated exon, and intracellular dialysis of bacterially expressed AMPK markedly attenuated BKCa currents in these cells. Conditional regulation of BKCa channel splice variants by AMPK may therefore determine the response of carotid body type I cells to hypoxia.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Corpo Carotídeo/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Animais Recém-Nascidos , Eletrofisiologia , Células HEK293 , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Alta/genética , Fígado/metabolismo , Camundongos , Fosforilação , Isoformas de Proteínas/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Adv Exp Med Biol ; 758: 81-90, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23080146

RESUMO

Our recent investigations provide further support for the proposal that, consequent to inhibition of mitochondrial oxidative phosphorylation, activation of AMP-activated protein kinase (AMPK) mediates carotid body excitation by hypoxia. Consistent with the effects of hypoxia, intracellular dialysis from a patch pipette of an active (thiophosphorylated) recombinant AMPK heterotrimer (α2ß2γ1) or application of the AMPK activators AICAR and A769662: (1) Inhibited BK(Ca) currents and TASK K(+) currents in rat carotid body type I cells; (2) Inhibited whole-cell currents carried by KCa1.1 and TASK3, but not TASK1 channels expressed in HEK293 cells; (3) Triggered carotid body activation. Furthermore, preliminary studies using mice with conditional knockout in type I cells of the primary upstream kinase that activates AMPK in response to metabolic stresses, LKB1, appear to confirm our working hypothesis. Studies on mice with knockout of the catalytic α1 subunit and α2 subunits of AMPK, respectively, have proved equally consistent. Accumulating evidence therefore suggests that the LKB1-AMPK signalling pathway is necessary for hypoxia-response coupling by the carotid body, and serves to regulate oxygen and therefore energy supply at the whole body level.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Corpo Carotídeo/fisiologia , Homeostase , Hipóxia/fisiopatologia , Canais de Potássio/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Transdução de Sinais/fisiologia , Quinases Proteína-Quinases Ativadas por AMP , Animais , Humanos , Proteínas do Tecido Nervoso/fisiologia , Canais de Potássio de Domínios Poros em Tandem/fisiologia
10.
Front Physiol ; 13: 874039, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35510145

RESUMO

The Carotid Bodies (CB) are peripheral chemoreceptors that detect changes in arterial oxygenation and, via afferent inputs to the brainstem, correct the pattern of breathing to restore blood gas homeostasis. Herein, preliminary evidence is presented supporting a novel oxygen-sensing hypothesis which suggests CB Type I cell "hypoxic signaling" may in part be mediated by mitochondria-generated thermal transients in TASK-channel-containing microdomains. Distances were measured between antibody-labeled mitochondria and TASK-potassium channels in primary rat CB Type I cells. Sub-micron distance measurements (TASK-1: 0.33 ± 0.04 µm, n = 47 vs TASK-3: 0.32 ± 0.03 µm, n = 54) provided evidence for CB Type I cell oxygen-sensing microdomains. A temperature-sensitive dye (ERthermAC) indicated that inhibition of mitochondrial activity in isolated cells caused a rapid and reversible inhibition of mitochondrial thermogenesis and thus temperature in these microdomains. Whole-cell perforated-patch current-clamp electrophysiological recordings demonstrated sensitivity of resting membrane potential (Vm) to temperature: lowering bath temperature from 37°C to 24°C induced consistent and reversible depolarizations (Vm at 37°C: -48.4 ± 4.11 mV vs 24°C: -31.0 ± 5.69 mV; n = 5; p < 0.01). These data suggest that hypoxic inhibition of mitochondrial thermogenesis may play an important role in oxygen chemotransduction in the CB. A reduction in temperature within cellular microdomains will inhibit plasma membrane ion channels, influence the balance of cellular phosphorylation-dephosphorylation, and may extend the half-life of reactive oxygen species. The characterization of a thermosensory chemotransduction mechanism, that may also be used by other oxygen-sensitive cell types and may impact multiple other chemotransduction mechanisms is critical if we are to fully understand how the CBs, and potentially other oxygen-sensitive cells, respond to hypoxia.

11.
Cell Calcium ; 44(2): 190-201, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18191199

RESUMO

In arterial myocytes the Ca(2+) mobilizing messenger NAADP evokes spatially restricted Ca(2+) bursts from a lysosome-related store that are subsequently amplified into global Ca(2+) waves by Ca(2+)-induced Ca(2+)-release from the sarcoplasmic reticulum (SR) via ryanodine receptors (RyRs). Lysosomes facilitate this process by forming clusters that co-localize with a subpopulation of RyRs on the SR. We determine here whether RyR subtypes 1, 2 or 3 selectively co-localize with lysosomal clusters in pulmonary arterial myocytes using affinity purified specific antibodies. The density of: (1) alphalgP120 labelling, a lysosome-specific protein, in the perinuclear region of the cell (within 1.5mum of the nucleus) was approximately 4-fold greater than in the sub-plasmalemmal (within 1.5mum of the plasma membrane) and approximately 2-fold greater than in the extra-perinuclear (remainder) regions; (2) RyR3 labelling within the perinuclear region was approximately 4- and approximately 14-fold greater than that in the extra-perinuclear and sub-plasmalemmal regions, and approximately 2-fold greater than that for either RyR1 or RyR2; (3) despite there being no difference in the overall densities of fluorescent labelling of lysosomes and RyR subtypes between cells, co-localization with alphalgp120 labelling within the perinuclear region was approximately 2-fold greater for RyR3 than for RyR2 or RyR1; (4) co-localization between alphalgp120 and each RyR subtype declined markedly outside the perinuclear region. Furthermore, selective block of RyR3 and RyR1 with dantrolene (30muM) abolished global Ca(2+) waves but not Ca(2+) bursts in response to intracellular dialysis of NAADP (10nM). We conclude that a subpopulation of lysosomes cluster in the perinuclear region of the cell and form junctions with SR containing a high density of RyR3 to comprise a trigger zone for Ca(2+) signalling by NAADP.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Pulmão/metabolismo , Lisossomos/metabolismo , Músculo Liso Vascular/metabolismo , NADP/análogos & derivados , Artéria Pulmonar/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Animais , Células Cultivadas , Fluorescência , Coração/fisiologia , Pulmão/citologia , Masculino , Músculo Liso Vascular/citologia , NADP/metabolismo , Isoformas de Proteínas , Artéria Pulmonar/citologia , Ratos , Ratos Wistar , Rianodina/farmacologia , Retículo Sarcoplasmático/metabolismo
12.
Eur J Pharmacol ; 595(1-3): 39-43, 2008 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-18703047

RESUMO

Hypoxic pulmonary vasoconstriction is a vital homeostatic mechanism that aids ventilation-perfusion matching in the lung, for which the underlying mechanism(s) remains controversial. However, our most recent investigations strongly suggest that hypoxic pulmonary vasoconstriction is precipitated, at least in part, by the inhibition of mitochondrial oxidative phosphorylation by hypoxia, an increase in the AMP/ATP ratio and consequent activation of AMP-activated protein kinase (AMPK). Unfortunately, these studies lacked the definitive proof that can only be provided by selectively blocking AMPK-dependent signalling cascades. The aim of the present study was, therefore, to determine the effects of the AMPK inhibitor compound C upon: (1) phosphorylation in response to hypoxia of a classical AMPK substrate, acetyl CoA carboxylase, in rat pulmonary arterial smooth muscle and (2) hypoxic pulmonary vasoconstriction in rat isolated intrapulmonary arteries. Acetyl CoA carboxylase phosphorylation was increased approximately 3 fold in the presence of hypoxia (pO(2) = 16-21 mm Hg, 1 h) and 5-aminoimidazole-4-carboxamide riboside (AICAR; 1 mM; 4 h) and in a manner that was significantly attenuated by the AMPK antagonist compound C (40 microM). Most importantly, pre-incubation of intrapulmonary arteries with compound C (40 microM) inhibited phase II, but not phase I, of hypoxic pulmonary vasoconstriction. Likewise, compound C (40 microM) inhibited constriction by AICAR (1 mM). The results of the present study are consistent with the activation of AMPK being a key event in the initiation of the contractile response of pulmonary arteries to acute hypoxia.


Assuntos
Hipóxia/enzimologia , Complexos Multienzimáticos/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Artéria Pulmonar/efeitos dos fármacos , Pirazóis/farmacologia , Pirimidinas/farmacologia , Vasoconstrição/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP , Acetil-CoA Carboxilase/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Cálcio/metabolismo , Dinoprosta/metabolismo , Hipóxia/fisiopatologia , Masculino , Complexos Multienzimáticos/metabolismo , Fosforilação , Potássio/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Artéria Pulmonar/enzimologia , Ratos , Ratos Sprague-Dawley , Ribonucleotídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos
13.
Adv Exp Med Biol ; 605: 63-8, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18085248

RESUMO

The carotid bodies play a critical role in initiating compensatory ventilatory responses to hypoxia. However, the complete mechanism by which hypoxia excites the oxygen-sensing carotid body type 1 cells has not been fully defined. We have previously proposed that the enzyme adenosine monophosphate-activated protein kinase (AMPK) may couple hypoxic inhibition of mitochondrial oxidative phosphorylation to carotid body type I cell excitation (Evans, Mustard, Wyatt, Peers, Dipp, Kumar, Kinnear and Hardie 2005). Here we discuss evidence that AMPK is a key requirement for hypoxic chemotransduction by the carotid body. In addition, we postulate upon a role for AMPK in the plasticity observed in the carotid body during both chronic and chronic intermittent hypoxia.


Assuntos
Corpo Carotídeo/fisiologia , Complexos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Ativadas por AMP , Animais , Animais Recém-Nascidos , Corpo Carotídeo/enzimologia , Hipóxia/fisiopatologia , Microcirculação/fisiologia , Subunidades Proteicas/metabolismo , Ratos
14.
Nat Commun ; 9(1): 4030, 2018 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-30279412

RESUMO

Asthma accounts for 380,000 deaths a year. Carotid body denervation has been shown to have a profound effect on airway hyper-responsiveness in animal models but a mechanistic explanation is lacking. Here we demonstrate, using a rat model of asthma (OVA-sensitized), that carotid body activation during airborne allergic provocation is caused by systemic release of lysophosphatidic acid (LPA). Carotid body activation by LPA involves TRPV1 and LPA-specific receptors, and induces parasympathetic (vagal) activity. We demonstrate that this activation is sufficient to cause acute bronchoconstriction. Moreover, we show that prophylactic administration of TRPV1 (AMG9810) and LPA (BrP-LPA) receptor antagonists prevents bradykinin-induced asthmatic bronchoconstriction and, if administered following allergen exposure, reduces the associated respiratory distress. Our discovery provides mechanistic insight into the critical roles of carotid body LPA receptors in allergen-induced respiratory distress and suggests alternate treatment options for asthma.


Assuntos
Acrilamidas/uso terapêutico , Asma/prevenção & controle , Compostos Bicíclicos Heterocíclicos com Pontes/uso terapêutico , Corpo Carotídeo/metabolismo , Lisofosfolipídeos/uso terapêutico , Receptores de Ácidos Lisofosfatídicos/metabolismo , Canais de Cátion TRPV/metabolismo , Acrilamidas/farmacologia , Animais , Asma/etiologia , Asma/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Lisofosfolipídeos/farmacologia , Masculino , Ratos Endogâmicos BN , Ratos Sprague-Dawley , Receptores de Ácidos Lisofosfatídicos/antagonistas & inibidores , Canais de Cátion TRPV/antagonistas & inibidores
15.
Respir Physiol Neurobiol ; 157(1): 75-82, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17157084

RESUMO

MaxiK channels are a unique class of K(+) channels activated by both voltage and intracellular Ca(2+). Derived from a single gene, their diversity arises from extensive splicing, and their wide distribution has led to their implication in a large variety of cellular functions. In the carotid body, they have been proposed to contribute to the resting membrane potential of type I cells, and also to be O(2) sensitive. Thus, they have been suggested to have an important role in hypoxic chemotransduction. Their O(2) sensitivity is preserved when the channels are expressed in HEK 293 cells, permitting detailed studies of candidate mechanisms underlying hypoxic inhibition of maxiK channels. In this article, we review evidence for and against an important role for maxiK channels in chemotransduction. We also consider different mechanisms proposed to account for hypoxic channel inhibition and suggest that, although our understanding of this important physiological process has advanced significantly in recent years, there remain important, unanswered questions as to the importance of maxiK in carotid body chemoreception.


Assuntos
Corpo Carotídeo/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Mecanotransdução Celular/fisiologia , Animais , Humanos , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/química
16.
Respir Physiol Neurobiol ; 157(1): 22-9, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17409030

RESUMO

AMP-activated protein kinase (AMPK) is a key component of a kinase cascade that regulates energy balance at the cellular level. Our recent research has raised the possibility that AMPK may also function to couple hypoxic inhibition of mitochondrial oxidative phosphorylation to O(2)-sensitive K(+) channel inhibition and hence underpin carotid body type I cell excitation. Thus, in addition to maintaining the cellular energy state AMPK may act as the primary metabolic sensor and effector of hypoxic chemotransduction in type I cells. These findings provide a unifying link between two previously separate theories pertaining to O(2)-sensing in the carotid body, namely the 'membrane hypothesis' and the 'mitochondrial hypothesis'. Furthermore, our data suggest that in addition to its effects at the cellular level the AMPK signalling cascade can mediate vital physiological mechanisms essential for meeting the metabolic needs of the whole organism.


Assuntos
Corpo Carotídeo/fisiologia , Mecanotransdução Celular/fisiologia , Modelos Biológicos , Complexos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/fisiologia , Proteínas Quinases Ativadas por AMP , Animais , Membrana Celular/metabolismo , Humanos , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Mitocôndrias/metabolismo , Canais de Potássio/metabolismo
18.
Stem Cells Dev ; 26(11): 808-817, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28335680

RESUMO

Placental abnormalities can cause Pregnancy-Associated Disorders, including preeclampsia, intrauterine growth restriction, and placental insufficiency, resulting in complications for both the mother and fetus. Trophoblast cells within the labyrinthine layer of the placenta facilitate the exchange of nutrients, gases, and waste between mother and fetus; therefore, the development of this cell layer is critical for fetal development. As trophoblast cells differentiate, it is assumed their metabolism changes with their energy requirements. We hypothesize that proper regulation of trophoblast metabolism is a key component of normal placental development; therefore, we examined the role of AMP-activated kinase (AMPK, PRKAA1/2), a sensor of cellular energy status. Our previous studies have shown that AMPK knockdown alters both trophoblast differentiation and nutrient transport. In this study, AMPKα1/2 shRNA was used to investigate the metabolic effects of AMPK knockdown on SM10 placental labyrinthine progenitor cells before and after differentiation. Extracellular flux analysis confirmed that AMPK knockdown was sufficient to reduce trophoblast glycolysis, mitochondrial respiration, and ATP coupling efficiency. A reduction in AMPK in differentiated trophoblasts also resulted in increased mitochondrial volume. These data indicate that a reduction in AMPK disrupts cellular metabolism in both progenitors and differentiated placental trophoblasts. This disruption correlates to abortive trophoblast differentiation that may contribute to the development of Pregnancy-Associated Disorders.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Diferenciação Celular , Vilosidades Coriônicas/metabolismo , Metabolismo Energético , Técnicas de Silenciamento de Genes , Células-Tronco/citologia , Células-Tronco/enzimologia , Trifosfato de Adenosina/metabolismo , Animais , Linhagem Celular , Respiração Celular , Forma Celular , Tamanho Celular , Feminino , Glicólise , Camundongos , Mitocôndrias/metabolismo , Tamanho das Organelas , Gravidez , Prótons
19.
Pharmacol Ther ; 107(3): 286-313, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16005073

RESUMO

It is generally accepted that the mobilisation of intracellular Ca2+ stores plays a pivotal role in the regulation of arterial smooth muscle function, paradoxically during both contraction and relaxation. However, the spatiotemporal pattern of different Ca2+ signals that elicit such responses may also contribute to the regulation of, for example, differential gene expression. These findings, among others, demonstrate the importance of discrete spatiotemporal Ca2+ signalling patterns and the mechanisms that underpin them. Of fundamental importance in this respect is the realisation that different Ca2+ storing organelles may be selected by the discrete or coordinated actions of multiple Ca2+ mobilising messengers. When considering such messengers, it is generally accepted that sarcoplasmic reticulum (SR) stores may be mobilised by the ubiquitous messenger inositol 1,4,5 trisphosphate. However, relatively little attention has been paid to the role of Ca2+ mobilising pyridine nucleotides in arterial smooth muscle, namely, cyclic adenosine diphosphate-ribose (cADPR) and nicotinic acid adenine dinucleotide phosphate (NAADP). This review will therefore focus on these novel mechanisms of calcium signalling and their likely therapeutic potential.


Assuntos
ADP-Ribose Cíclica/fisiologia , NADP/análogos & derivados , Sinalização do Cálcio , ADP-Ribose Cíclica/biossíntese , ADP-Ribose Cíclica/farmacologia , Humanos , Músculo Liso Vascular , NADP/biossíntese , NADP/farmacologia , NADP/fisiologia , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia , Retículo Sarcoplasmático/fisiologia , Vasoconstrição/fisiologia , Vasodilatação/fisiologia
20.
Novartis Found Symp ; 272: 73-85; discussion 85-94, 131-40, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16686430

RESUMO

The carotid body plays an important role in initiating protective responses to hypoxemia. The primary oxygen sensing cells are the glomus or type 1 cells. Hypoxia evokes the secretion of neurotransmitters from these cells which then excite afferent nerves. This response is mediated via membrane depolarization and voltage-gated Ca2+ entry. Studies from this laboratory have revealed that membrane depolarization in response to hypoxia is primarily the result of inhibition of background K+ channels which show strong similarities to the acid sensitive tandem-P-domain K+ channels TASK-1 and TASK-3. The background K+ channels of type-1 cells are also very sensitive to inhibition of mitochondrial energy metabolism and, in excised patches, appear to be directly activated by ATP. Thus these TASK-like background channels would appear to confer the ability to sense changes in oxygen levels, pH and metabolism upon the type 1 cell. The key issue of whether the effects of hypoxia are mediated through changes in metabolism remains unanswered but the effects of inhibition of mitochondrial energy metabolism and of hypoxia upon background K+ channels is mutually exclusive suggesting that there is a close link between metabolism and oxygen sensing in the type 1 cell.


Assuntos
Corpo Carotídeo/fisiologia , Oxigênio/metabolismo , Canais de Potássio de Domínios Poros em Tandem/fisiologia , Canais de Potássio/fisiologia , Animais , Sinalização do Cálcio/fisiologia , Corpo Carotídeo/metabolismo , Proteínas do Tecido Nervoso , Canais de Potássio/genética , Canais de Potássio/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA