首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 0 毫秒
1.
Intravascular CaCl2 infusion in freshwater rainbow trout (Salmo gairdneri) causes a significant degranulation of the corpuscles of Stannius (CS). Concurrently, there is a specific and acute inhibition of whole body Ca2+ influx; Ca2+ efflux is not effected. The material released from the CS after CaCl2 injection consists primarily of a 28-kDa product which we identified as hypocalcin. Electron microscope observations of the CS reveal that type 1 and type 2 cells are degranulated to a similar extent. We conclude that hypocalcin is directly involved in hypocalcemic control in freshwater fish via inhibition of branchial Ca2+ influx, thereby promoting a net loss of Ca2+ across the gill.  相似文献   

2.
Voltage-gated Ca2+ channels are categorized as either high-voltage activated (HVA) or low-voltage activated (LVA), and a subtype (or subtypes) of HVA Ca2+ channels link the presynaptic depolarization to rapid neuro-transmitter release. Reductions in transmitter release are characteristic of the autoimmune disorder, Lambert-Eaton syndrome (LES). Because antibodies from LES patients reduce Ca2+ influx in a variety of cell types and disrupt the intramembrane organization of active zones at neuromuscular synapses, specificity of LES antibodies for the Ca2+ channels that control transmitter release has been suggested as the mechanism for disease. We tested sera from four patients with LES. Serum samples from three of the four patients reduced both the maximal LVA and HVA Ca2+ conductances in murine dorsal root ganglion neurons. Thus, even though LES is expressed as a neuromuscular and autonomic disorder, our studies suggest that Ca2+ channels may be broadly affected in LES patients. To account for the specificity of disease expression, we suggest that incapacitation of only a fraction of the Ca2+ channels clustered at active zones would severely depress transmitter release. In particular, if several Ca2+ channels in a cluster are normally required to open simultaneously before transmitter release becomes likely, the loss of a few active zone Ca2+ channels would exponentially reduce the probability of transmitter release. This model may explain why LES is expressed as a neuromuscular disorder and can account for a clinical hallmark of LES, facilitation of neuromuscular transmission produced by vigorous voluntary effort.  相似文献   

3.
Zinc (Zn2+) is an essential metal in biology, and its bioavailability is highly regulated. Many cell types exhibit fluctuations in Zn2+ that appear to play an important role in cellular function. However, the detailed molecular mechanisms by which Zn2+ dynamics influence cell physiology remain enigmatic. Here, we use a combination of fluorescent biosensors and cell perturbations to define how changes in intracellular Zn2+ impact kinase signaling pathways. By simultaneously monitoring Zn2+ dynamics and kinase activity in individual cells, we quantify changes in labile Zn2+ and directly correlate changes in Zn2+ with ERK and Akt activity. Under our experimental conditions, Zn2+ fluctuations are not toxic and do not activate stress-dependent kinase signaling. We demonstrate that while Zn2+ can nonspecifically inhibit phosphatases leading to sustained kinase activation, ERK and Akt are predominantly activated via upstream signaling and through a common node via Ras. We provide a framework for quantification of Zn2+ fluctuations and correlate these fluctuations with signaling events in single cells to shed light on the role that Zn2+ dynamics play in healthy cell signaling.

Zinc (Zn2+) is an essential metal in biology, with approximately 10% of the proteins encoded by the human genome predicted to bind Zn2+ (1). All cells maintain and regulate a small pool of labile Zn2+ that can be exchanged among Zn2+-binding proteins and Zn2+ biosensors. The concentration of labile Zn2+ in the cytosol, measured in the hundreds of picomolar range (25), falls within the affinity range of many Zn2+ binding proteins, suggesting that under normal conditions many of these proteins will bind Zn2+ and function properly. However, some Zn2+ binders may need higher Zn2+ concentrations in order to function (6). Furthermore, there is growing evidence that mammalian cells experience fluctuations in available Zn2+, and these dynamics have been shown to be important for cell physiology (711).In addition to serving as an important biological cofactor (12), there are increasing examples that Zn2+ also plays a role in biological signaling. Crosstalk has been observed between Zn2+ dynamics and calcium signaling where increases in cytosolic Zn2+ lead to decrease in endoplasmic reticulum (ER) calcium, and conversely, increases in cytosolic calcium change Zn2+ homeostasis in the ER3. Zn2+ sequestration has been shown to block cell cycle progression in both meiotic oocytes (13) and mitotic cells (1416). At a molecular level, picomolar concentrations of Zn2+ potentiate the response of the ryanodine receptor in cardiomyocytes (17). Zn2+ has also been implicated in metabotropic signaling via the G protein–coupled receptor 39 (GPR39 (18)), direct modulation of protein kinase C activity (19), and activation of MAPK kinase signaling pathways in neurons (20), cardiomyocytes (21), and mast cells (7). While the above studies demonstrate that Zn2+ fluctuations influence cellular processes, in many cases the molecular details of how Zn2+ interacts with canonical signaling pathways, second messengers, or serves as a signal itself are unclear. This is especially true for the MAPK pathway.MAPK signaling plays a role in cell proliferation, differentiation, and development and is one of the most well-studied signaling pathways (22). A connection between MAPK signaling and Zn2+ was first reported in 1996 when it was observed that addition of 300 μM ZnCl2 to 3T3 fibroblasts led to increased phosphorylation of ERK1/2 kinases in the MAPK pathway (23). Early studies used epithelial cell lines to study the connection between Zn2+ and ERK signaling (23, 24). More recently, Zn2+ elevation has been demonstrated to increase ERK phosphorylation in dissociated neurons and transformed HT22 cells, where ERK signaling has been linked to synaptic plasticity and memory consolidation (20, 25, 26). The mechanism of ERK activation by Zn2+ remains enigmatic. The leading hypothesis has been that Zn2+ inhibits protein phosphatases, leading to sustained ERK activation. This idea is supported by the observation that ERK-directed phosphatase PP2A activity is reduced when Zn2+ is added to cell lysates (20, 25). Furthermore, it has been demonstrated that certain phosphatases are inhibited by nano- and picomolar concentrations of Zn2+ in vitro, although these phosphatases are not known to directly interact with ERK1/2 (27, 28). However, it is unclear how these bulk in vitro analyses relate to the role of Zn2+ fluctuations in living cells.The connection between Zn2+ and modulation of the MAPK pathway is even more perplexing when examining how Zn2+ influences Ras activity, which acts upstream of Raf-MEK-ERK. Two studies that involved acute perturbation of Zn2+ by adding high concentrations of Zn2+ concluded that Zn2+ promotes Ras activation (29, 30). On the other hand, two genetic screens in Caenorhabditis elegans suggested that Zn2+ inhibits Ras activity (31, 32). While these studies involved different model systems (cell lines versus C. elegans) and different means of altering Zn2+ (acute elevation versus chronic manipulation), it is important to note that there is a lack of consensus on how Zn2+ influences the Ras-Raf-MEK-ERK pathway.In this work we set out to dissect the connection between Zn2+ and ERK in an effort to elucidate the mechanism of activation. Using a combination of kinase translocation reporters and a Förster resonance energy transfer (FRET)-sensor for Zn2+, we quantified the changes in intracellular Zn2+ in response to subtle extracellular perturbations and correlated them directly with changes in kinase activity at the single cell level. We found that while elevated Zn2+ broadly inhibits phosphatase activity to some extent in vitro, in live cells, Zn2+ primarily activates ERK via upstream signaling, suggesting that ERK phosphatase inhibition can’t fully account for the Zn2+-induced increase in ERK activity. Finally, we demonstrate that our Zn2+ conditions activate Ras and Akt signaling along with ERK but that few other kinases are activated, including stress-response kinases JNK, p38, and p53. We therefore propose a mechanism of action where Zn2+ activates ERK and Akt pathways upstream of Ras, while the specific Zn2+-protein interaction remains elusive.  相似文献   

4.
OBJECTIVE: Conditions of ischemia-reperfusion disturb the homoeostasis of cytosolic Ca2+ in cardiac microvascular endothelial cells (CMEC), leading to numerous malfunctions of the endothelium. Reperfusion specifically aggravates the Ca2+ overload developed during sustained ischemia. The aim of this study was to identify the origin of the reperfusion-induced part of the Ca2+ overload. Our hypotheses were that this is either due to a Na+-dependent process, e.g. involving the Na+/H+ exchanger (NHE) and/or the Na+/Ca2+ exchanger (NCX), or a process involving the endoplasmic reticulum (ER) and store-operated channels (SOC). METHODS AND RESULTS: Cultured CMEC from rats were exposed to conditions of simulated ischemia (hypoxia, pH 6.4) and reperfusion (reoxygenation, pH 7.4). Cytosolic Ca2+ ([Ca2+]i) and cytosolic Na+ ([Na+]i) concentrations and cytosolic pH (pHi) were measured with the use of fluorescent indicators. Removal of Ca2+ from the extracellular media during reoxygenation prevented the [Ca2+]i rise. Neither the activation of the NHE nor of the NCX in reoxygenated CMEC caused a change in this [Ca2+]i rise. Complete or partial removal of Na+ from the external media also had no effect on the [Ca2+]i rise. In contrast, specific inhibition of the inositol trisphosphate (InsP3) receptor by xestospongin C (3 micromol/l), of phospholipase (PLC) by U73122 (1 micromol/l), or of SOC by the inhibitors gadolinium (10 micromol/l) or 2-APB (50 micromol/l) lowered or abolished the reoxygenation-induced [Ca2+]i rise. CONCLUSION: In CMEC exposed to reperfusion conditions, the enhanced Ca2+ overload is due to Ca2+ influx. The influx is not mediated by a Na+-dependent mechanism, but rather is due to activation of the InsP3 receptor of the ER and activation of SOC.  相似文献   

5.
Ca(2+) mobilizing agonists and hemodynamic shear stress both elicit a rise in endothelial cytosolic Ca(2+) [Ca(2+)](i), which then acts to stimulate nitric oxide synthase and phospholipase A(2), leading to the production and release of nitric oxide (NO) and other vascular substances such as prostacyclin and endothelium-derived hyperpolarizing factors (EDHF). In this article, regulatory mechanisms of agonist-induced and mechanosensitive Ca(2+) influx pathways in vascular endothelial cells will be discussed. Special emphasis will be placed on the regulation of agonist-induced Ca(2+) influx by protein kinase G (PKG). Flow-induced Ca(2+) influx in relation to vascular dilation and the vasodilator produced will also be discussed.  相似文献   

6.
7.
The endocardial endothelium is an important modulator of myocardial function. The present study demonstrates the existence of a stretch-activated Ca(2+)-permeable cation channel and of a Ca(2+)-activated K+ channel in the endocardial endothelium of the porcine right atrium. The stretch-activated channel is permeable for K+, Na+, Ca2+, and Ba2+, with mean conductances of approximately 32 pS for the monovalent cations and approximately 13 pS for divalent cations. The Ca(2+)-activated K+ channel has a mean conductance of 192 pS in symmetrical KCl. solution. Channel activity is strongly dependent on membrane potential and the cytosolic Ca2+ concentration. Half-maximal activation occurs at a cytosolic Ca2+ concentration of approximately 5 microM. The influx of Ca2+ through the stretch-activated channel is sufficient to activate the Ca(2+)-activated K+ channel in cell-attached patches. Upon activation of the stretch-activated channel, the cytosolic Ca2+ concentration increases, at least locally, to values of approximately 0.5 microM, as deduced from the open probability of the Ca(2+)-dependent K+ channel that was activated simultaneously. The stretch-activated channels are capable of inducing an intracellular Ca2+ signal and may have a role as mechanosensors in the atrial endothelium, possibly activated by atrial overload.  相似文献   

8.
Kaneishi K  Sakuma Y  Kobayashi H  Kato M 《Endocrinology》2002,143(11):4210-4217
In GT1-7 cells, cAMP increases the intracellular Ca2+ concentration ([Ca2+](i)) through activation of the voltage-gated Ca2+ channels, thereby facilitating GnRH release. To activate these channels, the membrane potential must be depolarized. In the present study we hypothesize that cAMP depolarizes the cells by increasing the membrane Na+ permeability, as in the case of somatotrophs and pancreatic beta-cells. To examine this, we analyzed [Ca2+](i) and [Na+](i) in GT1-7 cells by an intracellular ion-imaging technique along with cAMP assay by RIA. Forskolin, a direct activator of adenylyl cyclase, increased [Ca2+](i) and [Na+](i) via cAMP formation. The forskolin-induced increase in [Ca2+](i) depended on the presence of Ca2+ and Na+ in the extracellular solution. This response was blocked by the voltage-gated Ca2+ channel blocker, nifedipine; the nonselective cation channel blocker, gadolinium (Gd3+); and the cyclic nucleotide-gated channel blocker, l-cis-diltiazem. In contrast, the forskolin-induced increase in [Na+](i) depended only on extracellular Na+, not on Ca2+. Gd3+ and l-cis-diltiazem also blocked the increase in [Na+](i). Furthermore, the forskolin-induced increase in GnRH release was blunted in both low Ca2+ and low Na+ media. The results indicate that cAMP increases the membrane Na+ permeability, probably through nonselective cation channels on GT1-7 cells, thereby promoting GnRH release.  相似文献   

9.
Rapid, nonclassical 17β-estradiol (E2) actions are thought to play an important role in the modulation of neuronal function. The present study addresses the intracellular signaling cascades involved in the rapid E2-induced phosphorylation of cAMP response element binding protein (CREB) in GnRH neurons. Administration of E2 to adult female mice resulted in the activation of ERK1/2 in GnRH neurons within 15 min. In vitro studies using pharmacological antagonists showed that ERK1/2 was essential for E2-induced CREB phosphorylation in GnRH neurons. Upstream to this, protein kinase A and calcium/calmodulin-dependent protein kinase type II, but not protein kinase C, were found to be necessary for E2-induced phosphorylation of ERK1/2. This rapid E2 signaling cascade in GnRH neurons was found to require both direct and indirect E2 actions. E2 failed to phosphorylate ERK1/2 and CREB in GnRH neuron-specific estrogen receptor β knockout mice in vivo. Equally, however, a cocktail of tetrodotoxin and γ-aminobutyric acid(A)/glutamate receptor antagonists also blocked E2-induced ERK1/2 phosphorylation in GnRH neurons in wild-type mice in vitro. Together, these observations indicate that E2 acts through calcium/calmodulin-dependent protein kinase type II and protein kinase A to rapidly phosphorylate ERK1/2, which then acts to phosphorylate CREB in adult female GnRH neurons. Intriguingly, these effects of E2 are dependent upon both direct ERβ mechanisms as well as indirect actions mediated by afferent inputs to GnRH neurons.  相似文献   

10.
The effect of long-chain free fatty acids on glucagon secretion from islet alpha-cells has been a controversial issue. This study examined direct effects of oleic acid (OA) on glucagon release from rat pancreatic islets and on cytoplasmic Ca(2+) concentration ([Ca(2+)](i)) in single alpha-cells by fura-2 fluorescence imaging. OA at 30 microM increased glucagon release from isolated islets in the presence of low (2.8 mM) and elevated (8.3 mM) glucose concentrations. OA at 6-10 microm concentration-dependently increased [Ca(2+)](i) in alpha-cells, irrespective of glucose concentrations (1.4, 2.8, and 8.3 mM). OA at 10 mum increased [Ca(2+)](i) in 90% of alpha-cells. OA-induced [Ca(2+)](i) increases were strongly inhibited by the endoplasmic reticulum Ca(2+)-pump inhibitors cyclopiazonic acid and thapsigargin and by 2-aminoethoxydiphenyl borate, the blocker of both inositol 1,4,5-trisphosphate receptors and store-operated Ca(2+) channels. Furthermore, the amplitude, but not incidence, of OA-induced [Ca(2+)](i) increases was reduced substantially by Ca(2+)-free conditions and mildly by an L-type Ca(2+) channel blocker, nitrendipine, and an ATP-sensitive K(+) channel activator, diazoxide. OA-induced glucagon release was also inhibited mildly by nitrendipine and strongly by 2-aminoethoxydiphenyl borate. These results indicate that OA glucose-independently stimulates glucagon release by increasing [Ca(2+)](i) in rat pancreatic alpha-cells and that the [Ca(2+)](i) increase is triggered by Ca(2+) release from endoplasmic reticulum and amplified by Ca(2+) influx possibly via store-operated channels and via voltage-dependent L-type Ca(2+) channels. The glucose-independent action of OA to stimulate glucagon release from alpha-cells may operate under hypoglycemic conditions when plasma free fatty acids levels are elevated, possibly playing a role in maintaining glucose metabolism.  相似文献   

11.
The vasodilating mechanisms of the K+ channel openers—cromakalim, pinacidil, nicorandil, KRN2391, and Ki4032—were examined by measurement of the cytoplasmic Ca2+ concentration ([Ca2+]i) using the fura-2 method in canine or porcine coronary arterial smooth muscle. The five K+ channel openers all produced a reduction of [Ca2+]i in 5 and 30 mM KCl physiological salt solution (PSS), the effects of which were antagonized by tetrabutylammonium (TBA) or glibenclamide, but failed to affect [Ca2+]i in 45 and 90 mM MCl-PSS. Cromakalim and Ki4032 only partially inhibited the 30 mM KCl-induced contractures, whereas pinacidil, nicorandil, and KRN2391 nearly abolished contractions produced by high KCl-PSS. The increased [Ca2+]i and force produced by a thromboxane A2 analogue, U46619, were inhibited by K+ channel openers and verapamil. In the absence of extracellular Ca2+, U46619 induced a transient increase in [Ca2+]i with a contraction, which is effectively inhibited by cromakalim and Ki4032. Their inhibitory effects were blocked by TBA and counteracted by 20 mM KCl-induced depolarization. Cromakalim and Ki4032 did not affect caffeine-induced Ca2+ release. Cromakalim reduced U46619-induced IP3 production and TBA blocked this inhibitory effect. Thus, cromakalim and Ki4032 are more specific K+ channel openers than pinacidil, nicorandil, and KRN2391. The vasodilation related with a reduction of [Ca2+]i produced by K+ channel openers is due to the hyperpolarization of the plasma membrane resulting in not only the closure of voltage-dependent Ca2+ channels but also inhibition of the production of IP3 and Ca2+ release from intracellular stores related to stimulation of the thromboxane A2 receptor.  相似文献   

12.
Whole-cell patch-clamp recordings of membrane currents and Fura-2 measurements of free intracellular calcium concentration ([Ca2+]i) were used to study calcium influx through receptor-activated cation channels in rat peritoneal mast cells. Cation channels were activated by the secretagogue compound 48/80, whereas a possible concomitant Ca2+ entry through pathways activated by depletion of calcium stores was blocked by dialyzing cells with heparin. Heparin effectively suppressed the transient Ca2+ release induced by 48/80 and abrogated inositol 1,4,5-trisphosphate-induced calcium influx without affecting activation of 50-pS cation channels. There was a clear correlation between changes in [Ca2+]i and the activity of 50-pS channels. The changes in [Ca2+]i increased with elevation of extracellular Ca2+. At the same time, inward currents through 50-pS channels were diminished as more Ca2+ permeated. This effect was due to a decrease in slope conductance and a reduction in the open probability of the cation channels. In physiological solutions, 3.6% of the total current was carried by Ca2+. The cation channels were not only permeable to Ca2+ but also to Mn2+, as evidenced by the quench of Fura-2 fluorescence. Mn2+ current through 50-pS channels could not be resolved at the single-channel level. Our results suggest that 50-pS cation channels partially contribute to sustained increases of [Ca2+]i in mast cells following receptor activation.  相似文献   

13.
Epinephrine, through its action on β-adrenoceptors, may induce endothelium-dependent vascular dilation, and this action is partly mediated by a cytosolic Ca2+ ([Ca2+]i) change in endothelial cells. In the present study, we explored the molecular identity of the channels that mediate epinephrine-induced endothelial Ca2+ influx and subsequent vascular relaxation. Patch clamp recorded an epinephrine- and cAMP-activated cation current in the primary cultured bovine aortic endothelial cells (BAECs) and H5V endothelial cells. L-cis-diltiazem and LY-83583, two selective inhibitors for cyclic nucleotide-gated channels, diminished this cation current. Furthermore, this cation current was greatly reduced by a CNGA2-specific siRNA in H5V cells. With the use of fluorescent Ca2+ dye, it was found that epinephrine and isoprenaline, a β-adrenoceptor agonist, induced endothelial Ca2+ influx in the presence of bradykinin. This Ca2+ influx was inhibited by L-cis-diltiazem and LY-83583, and by a β2-adrenoceptor antagonist ICI-118551. CNGA2-specific siRNA also diminished this Ca2+ influx in H5V cells. Furthermore, L-cis-diltiazem and LY-83583 inhibited the endothelial Ca2+ influx in isolated mouse aortic strips. L-cis-diltiazem also markedly reduced the endothelium-dependent vascular dilation to isoprenaline in isolated mouse aortic segments. In summary, CNG channels, CNGA2 in particular, mediate β-adrenoceptor agonist-induced endothelial Ca2+ influx and subsequent vascular dilation.  相似文献   

14.
Corpus striatum neurons acutely dissociated from the brains of young adult rats had membrane surfaces suitable for G omega-seal recording. Whole-cell current-clamp and voltage-clamp recordings indicated that the cells remained electrically excitable after dissociation. Cell-attached recordings frequently revealed single-channel openings in the presence of dopamine or of the D2 dopamine agonist quinpirole. Channel openings were rarely or never observed in the absence of drugs or in the presence of quinpirole plus the dopamine antagonist haloperidol. The D2 antagonist spiperone was more potent at blocking the appearance of the channel than was the D1 antagonist SCH-23390. The channel reversal potential varied with the extracellular K+ concentration as predicted by the Nernst equation. The channel current-voltage relationship was linear, with a conductance of approximately equal to 85 pS in the presence of 140 mM KCl. These results are consistent with the opening of single K+ channels following D2 dopamine receptor activation.  相似文献   

15.
To evaluate the role of Ca2+ influx on hormone secretion induced by cell swelling, we have utilized a prolactin (PRL)-secreting rat tumor cell line, MMQ, which has plasmalemma dopamine receptors. Medium hyposmolarity or osmotically equivalent isotonic urea caused prompt cell swelling and a rise in both [Ca2+]i and PRL secretion in a dose-dependent manner. Dopamine inhibited the induced increase in both [Ca2+]i and PRL secretion in a dose-dependent manner but the maximum inhibition was only 50%. This effect of dopamine was prevented by haloperidol. Depletion of medium Ca2+ or blocking Ca2+ influx with nifedipine completely abolished the osmotically induced rise in both [Ca2+]i and PRL secretion. These data indicate that Ca2+ influx through nifedipine-sensitive Ca2+ channels is an essential component of PRL secretion induced by osmotic cell swelling in MMQ cells and that a dopaminergic receptor-linked mechanism influences the opening of these channels.  相似文献   

16.
The application of confocal microscopy to cardiac and skeletal muscle has resulted in the observation of transient, spatially localized elevations in [Ca2+]i, termed 'Ca2+ sparks'. Ca2+ sparks are thought to represent 'elementary' Ca2+ release events, which arise from one or more ryanodine receptor (RyR) channels in the sarcoplasmic reticulum. In cardiac muscle, Ca2+ sparks appear to be key elements of excitation-contraction coupling, in which the global [Ca2+]i transient is thought to involve the recruitment of Ca2+ sparks, each of which is controlled locally by single coassociated L-type Ca2+ channels. Recently, Ca2+ sparks have been detected in smooth muscle cells of arteries. In this review, we analyse the complex relationship of Ca2+ influx and Ca2+ release with local, subcellular Ca2+ microdomains in light of recent studies on Ca2+ sparks in cardiovascular cells. We performed a comparative analysis of 'elementary' Ca2+ release units in mouse, rat and human arterial smooth muscle cells, using measurements of Ca2+ sparks and plasmalemmal K(Ca) currents activated by Ca2+ sparks (STOCs). Furthermore, the appearance of Ca2+ sparks during ontogeny of arterial smooth muscle is explored. Using intact pressurized arteries, we have investigated whether RyRs causing Ca2+ sparks (but not smaller 'quantized' Ca2+ release events, e.g. hypothetical 'Ca2+ quarks') function as key signals that, through membrane potential and global cytoplasmic [Ca2+], oppose arterial myogenic tone and influence vasorelaxation. We believe that voltage-dependent Ca2+ channels and local RyR-related Ca2+ signals are important in differentiation, proliferation, and gene expression. Our findings suggest that 'elementary' Ca2+ release units may represent novel potent therapeutic targets for regulating function of intact arterial smooth muscle tissue.  相似文献   

17.
Molecules of astrocyte origin influence gonadotropin-releasing hormone (GnRH) release and GnRH neuronal growth and differentiation. Furthermore, type 1 astrocytes express steroid receptors, presenting the possibility that steroid actions on GnRH neurons might occur via astrocytes. Utilizing GT1-7 cells, a GnRH-secreting cell line, the present study demonstrates that astrocytes mediate dehydroepiandrosterone (DHEA) or estradiol (E2) stimulated GnRH secretion. Conditioned media (CM) from astrocytes cultured for 48 h alone, with DHEA (DHEA-CM), or with E2 (E2-CM) were collected, treated with charcoal to remove steroids, and added to GT1-7 cells in culture for 12 h to test the effect on GnRH secretion. DHEA-CM and E2-CM stimulated GnRH secretion by GT1-7 cells by 4- and 3-fold, respectively. The effect of DHEA-CM on GnRH secretion by GT1-7 cells appears to be related to both DHEA and its metabolite, E2, since blocking the metabolism of DHEA into estrogen in the DHEA-treated astrocytes partially reversed the stimulatory effect of DHEA-CM. Addition of transforming growth factor (TGF)-beta1-neutralizing antibody to the astrocyte cultures reversed the stimulatory effects of both DHEA-CM and E2-CM on GnRH secretion by GT1-7 cells, suggesting that TGF-beta1 derived from astrocytes may be the principle mediator of E2 and DHEA effects. These data provide evidence for a novel mechanism by which circulating steroids and/or neurosteroids may modulate GnRH secretion.  相似文献   

18.
T-type Ca(2+) channels (TTCCs) are expressed in the developing heart, are not present in the adult ventricle, and are reexpressed in cardiac diseases involving cardiac dysfunction and premature, arrhythmogenic death. The goal of this study was to determine the functional role of increased Ca(2+) influx through reexpressed TTCCs in the adult heart. A mouse line with cardiac-specific, conditional expression of the alpha1G-TTCC was used to increase Ca(2+) influx through TTCCs. alpha1G hearts had mild increases in contractility but no cardiac histopathology or premature death. This contrasts with the pathological phenotype of a previously studied mouse with increased Ca(2+) influx through the L-type Ca(2+) channel (LTCC) secondary to overexpression of its beta2a subunit. Although alpha1G and beta2a myocytes had similar increases in Ca(2+) influx, alpha1G myocytes had smaller increases in contraction magnitude, and, unlike beta2a myocytes, there were no increases in sarcoplasmic reticulum Ca(2+) loading. Ca(2+) influx through TTCCs also did not induce normal sarcoplasmic reticulum Ca(2+) release. alpha1G myocytes had changes in LTCC, SERCA2a, and phospholamban abundance, which appear to be adaptations that help maintain Ca(2+) homeostasis. Immunostaining suggested that the majority of alpha1G-TTCCs were on the surface membrane. Osmotic shock, which selectively eliminates T-tubules, induced a greater reduction in L- versus TTCC currents. These studies suggest that T- and LTCCs are in different portions of the sarcolemma (surface membrane versus T-tubules) and that Ca(2+) influx through these channels induce different effects on myocyte contractility and lead to distinct cardiac phenotypes.  相似文献   

19.

Aims/hypothesis

There is evidence that ATP acts as an autocrine signal in beta cells but the receptors and pathways involved are incompletely understood. Here we investigate the receptor subtype(s) and mechanism(s) mediating the effects of ATP on human beta cells.

Methods

We examined the effects of purinergic agonists and antagonists on membrane potential, membrane currents, intracellular Ca2+ ([Ca2+]i) and insulin secretion in human beta cells.

Results

Extracellular application of ATP evoked small inward currents (3.4?±?0.7 pA) accompanied by depolarisation of the membrane potential (by 14.4?±?2.4 mV) and stimulation of electrical activity at 6 mmol/l glucose. ATP increased [Ca2+]i by stimulating Ca2+ influx and evoking Ca2+ release via InsP3-receptors in the endoplasmic reticulum (ER). ATP-evoked Ca2+ release was sufficient to trigger exocytosis in cells voltage-clamped at ?70 mV. All effects of ATP were mimicked by the P2Y(1/12/13) agonist ADP and the P2Y1 agonist MRS-2365, whereas the P2X(1/3) agonist α,β-methyleneadenosine-5-triphosphate only had a small effect. The P2Y1 antagonists MRS-2279 and MRS-2500 hyperpolarised glucose-stimulated beta cells and lowered [Ca2+]i in the absence of exogenously added ATP and inhibited glucose-induced insulin secretion by 35%. In voltage-clamped cells subjected to action potential-like stimulation, MRS-2279 decreased [Ca2+]i and exocytosis without affecting Ca2+ influx.

Conclusions/interpretation

These data demonstrate that ATP acts as a positive autocrine signal in human beta cells by activating P2Y1 receptors, stimulating electrical activity and coupling Ca2+ influx to Ca2+ release from ER stores.  相似文献   

20.
The exchange factor directly activated by cAMP (Epac) is a newly discovered direct target for cAMP and a guanine-nucleotide exchange factor for the small GTPase Rap. Little is known about the neuronal functions of Epac. Here we show that activation of Epac by specific cAMP analogs or by the pituitary adenylate cyclase-activating polypeptide induces a potent activation of the Ca2+-sensitive big K+ channel, slight membrane hyperpolarization, and increased after-hyperpolarization in cultured cerebellar granule cells. These effects involve activation of Rap and p38 MAPK, which mobilizes intracellular Ca2+ stores. These findings reveal a cAMP Epac-dependent and protein kinase A-independent signaling cascade that controls neuronal excitability.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号