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 15 毫秒
1.
The liberation of calcium ions sequestered in the endoplasmic reticulum through inositol 1,4,5-trisphosphate receptors/channels (IP3Rs) results in a spatiotemporal hierarchy of calcium signaling events that range from single-channel openings to local Ca2+ puffs believed to arise from several to tens of clustered IP3Rs to global calcium waves. Using high-resolution confocal linescan imaging and a sensitive Ca2+ indicator dye (fluo-4-dextran), we show that puffs are often preceded by small, transient Ca2+ elevations that we christen “trigger events”. The magnitude of triggers is consistent with their arising from the opening of a single IP3 receptor/channel, and we propose that they initiate puffs by recruiting neighboring IP3Rs within the cluster by a regenerative process of Ca2+-induced Ca2+ release. Puff amplitudes (fluorescence ratio change) are on average ~6 times greater than that of the triggers, suggesting that at least six IP3Rs may simultaneously be open during a puff. Trigger events have average durations of ~12 ms, as compared to 19 ms for the mean rise time of puffs, and their spatial extent is ~3 times smaller than puffs (respective widths at half peak amplitude 0.6 and 1.6 μm). All these parameters were relatively independent of IP3 concentration, although the proportion of puffs showing resolved triggers was greatest (~80%) at low [IP3]. Because Ca2+ puffs constitute the building blocks from which cellular IP3-mediated Ca2+ signals are constructed, the events that initiate them are likely to be of fundamental importance for cell signaling. Moreover, the trigger events provide a useful yardstick by which to derive information regarding the number and spatial arrangement of IP3Rs within clusters.  相似文献   

2.
This study presents an investigation of pacemaker mechanisms underlying lymphatic vasomotion. We tested the hypothesis that active inositol 1,4,5-trisphosphate receptor (IP3R)-operated Ca2+ stores interact as coupled oscillators to produce near-synchronous Ca2+ release events and associated pacemaker potentials, this driving action potentials and constrictions of lymphatic smooth muscle. Application of endothelin 1 (ET-1), an agonist known to enhance synthesis of IP3, to quiescent lymphatic smooth muscle syncytia first enhanced spontaneous Ca2+ transients and/or intracellular Ca2+ waves. Larger near-synchronous Ca2+ transients then occurred leading to global synchronous Ca2+ transients associated with action potentials and resultant vasomotion. In contrast, blockade of L-type Ca2+ channels with nifedipine prevented ET-1 from inducing near-synchronous Ca2+ transients and resultant action potentials, leaving only asynchronous Ca2+ transients and local Ca2+ waves. These data were well simulated by a model of lymphatic smooth muscle with: 1), oscillatory Ca2+ release from IP3R-operated Ca2+ stores, which causes depolarization; 2), L-type Ca2+ channels; and 3), gap junctions between cells. Stimulation of the stores caused global pacemaker activity through coupled oscillator-based entrainment of the stores. Membrane potential changes and positive feedback by L-type Ca2+ channels to produce more store activity were fundamental to this process providing long-range electrochemical coupling between the Ca2+ store oscillators. We conclude that lymphatic pacemaking is mediated by coupled oscillator-based interactions between active Ca2+ stores. These are weakly coupled by inter- and intracellular diffusion of store activators and strongly coupled by membrane potential. Ca2+ store-based pacemaking is predicted for cellular systems where: 1), oscillatory Ca2+ release induces depolarization; 2), membrane depolarization provides positive feedback to induce further store Ca2+ release; and 3), cells are interconnected. These conditions are met in a surprisingly large number of cellular systems including gastrointestinal, lymphatic, urethral, and vascular tissues, and in heart pacemaker cells.  相似文献   

3.
In polarized epithelial cells, agonists trigger Ca2+ waves and oscillations. These patterns may be caused by the compartmentalization of inositol 1,4,5-trisphosphate (IP3)-sensitive Ca2+ pools into specific regions. We have investigated the relationship between the distribution of IP3 receptors (IP3Rs) and the spatiotemporal pattern of Ca2+ signaling in the duct cells of the rat submandibular gland (SMG). Using immunofluorescence, although labeling was somewhat heterogeneous, the IP3Rs were colocalized to the apical pole of the duct cells. Immunoelectron microscopy identified small apical vesicles bearing IP3R2 in some types of duct cells. Real-time confocal imaging of intact ducts demonstrated that, after carbachol stimulation, an initial Ca2+ spike occurred in the apical region. Subsequently, repetitive Ca2+ spikes spread from the apical to the middle cytoplasm. These apical Ca2+ initiation sites were found only in some “pioneer cells,” rather than in all duct cells. We performed both Ca2+ imaging and immunofluorescence on the same ducts and detected the strongest immunosignals of IP3R2 in the Ca2+ initiation sites of the pioneer cells. The subcellular localization and expression level of IP3Rs correlated strongly with the spatiotemporal nature of the intracellular Ca2+ signal and distinct Ca2+ responses among the rat SMG duct cells.  相似文献   

4.
Waves of elevated intracellular free Ca2+ that propagate between neighboring astrocytes (Ca2+ waves) are important for the communication among astrocytes. We have previously revealed that focal photolysis of a caged calcium ionophore results in an increase in the concentration of intracellular Ca2+ in the target astrocytes, then the increase propagates to neighboring astrocytes through gap junctions. The extracellular ATP-purinoceptors signaling pathways are not primarily responsible for the propagation of the photolytic flash-induced Ca2+ waves. Here we examined whether and if so how the dynamics of Ca2+ waves changed after treatment with sublethal simulated ischemia; oxygen-glucose deprivation (OGD). OGD treatment increased the astrocytic expression of P2Y1 and P2Y2 receptors early during reperfusion, resulting in an increase in the propagating waves speed. In contrast, the expression of a gap junction protein was not changed significantly by the OGD suggesting that the extracellular ATP-P2Y receptors signaling pathways were preferentially enhanced after OGD. The present method to induce Ca2+ waves by focal photolysis of a caged calcium ionophore may provide a valuable tool with which to analyze glial Ca2+ waves under not only normal but also pathologic conditions.  相似文献   

5.
Sustained elevation of intracellular calcium by Ca2+ release–activated Ca2+ channels is required for lymphocyte activation. Sustained Ca2+ entry requires endoplasmic reticulum (ER) Ca2+ depletion and prolonged activation of inositol 1,4,5-trisphosphate receptor (IP3R)/Ca2+ release channels. However, a major isoform in lymphocyte ER, IP3R1, is inhibited by elevated levels of cytosolic Ca2+, and the mechanism that enables the prolonged activation of IP3R1 required for lymphocyte activation is unclear. We show that IP3R1 binds to the scaffolding protein linker of activated T cells and colocalizes with the T cell receptor during activation, resulting in persistent phosphorylation of IP3R1 at Tyr353. This phosphorylation increases the sensitivity of the channel to activation by IP3 and renders the channel less sensitive to Ca2+-induced inactivation. Expression of a mutant IP3R1-Y353F channel in lymphocytes causes defective Ca2+ signaling and decreased nuclear factor of activated T cells activation. Thus, tyrosine phosphorylation of IP3R1-Y353 may have an important function in maintaining elevated cytosolic Ca2+ levels during lymphocyte activation.  相似文献   

6.
Interactions between cyclic adenosine monophosphate (cAMP) and Ca2+ are widespread, and for both intracellular messengers, their spatial organization is important. Parathyroid hormone (PTH) stimulates formation of cAMP and sensitizes inositol 1,4,5-trisphosphate receptors (IP3R) to IP3. We show that PTH communicates with IP3R via “cAMP junctions” that allow local delivery of a supramaximal concentration of cAMP to IP3R, directly increasing their sensitivity to IP3. These junctions are robust binary switches that are digitally recruited by increasing concentrations of PTH. Human embryonic kidney cells express several isoforms of adenylyl cyclase (AC) and IP3R, but IP3R2 and AC6 are specifically associated, and inhibition of AC6 or IP3R2 expression by small interfering RNA selectively attenuates potentiation of Ca2+ signals by PTH. We define two modes of cAMP signaling: binary, where cAMP passes directly from AC6 to IP3R2; and analogue, where local gradients of cAMP concentration regulate cAMP effectors more remote from AC. Binary signaling requires localized delivery of cAMP, whereas analogue signaling is more dependent on localized cAMP degradation.  相似文献   

7.
Networks of glial cells, and in particular astrocytes, are capable of sustaining calcium (Ca2+) waves both in vivo and in vitro. Experimentally, it has been shown that there are two separate modes of communication: the first by the passage of an agent (inositol 1,4,5-triphosphate, IP3) through gap junctions (GJs) joining cells; the second by the diffusion of an extracellular agent (adenosine triphosphate, ATP) that binds to receptors on the cells. In both cases, the outcome is the release of Ca2+ from internal stores in the glial cells. These two modes of communication are not mutually exclusive, but probably work in conjunction in many cases. We present a model of a two-dimensional network of glial cells that incorporates regenerative intercellular (GJ) and extracellular (ATP) pathways. In the extreme cases of only one type of pathway, the results are in agreement with previous models. Adding an extracellular pathway to the GJ model increased the extent and duration of the Ca2+ wave, but did not significantly change the speed of propagation. Conversely, adding GJs to the extracellular model did increase the wave speed. The model was modified to apply to the retina by extending it to include both astrocytes and Müller cells, with GJs the dominant coupling between astrocytes and ATP responsible for most of the remaining communication. It was found that both pathways are necessary to account for experimental results.  相似文献   

8.
Disrupting inositol 1,4,5-trisphosphate (IP3) receptor (IP3R)/B-cell lymphoma 2 (Bcl-2) complexes using a cell-permeable peptide (stabilized TAT-fused IP3R-derived peptide (TAT-IDPS)) that selectively targets the BH4 domain of Bcl-2 but not that of B-cell lymphoma 2-extra large (Bcl-Xl) potentiated pro-apoptotic Ca2+ signaling in chronic lymphocytic leukemia cells. However, the molecular mechanisms rendering cancer cells but not normal cells particularly sensitive to disrupting IP3R/Bcl-2 complexes are poorly understood. Therefore, we studied the effect of TAT-IDPS in a more heterogeneous Bcl-2-dependent cancer model using a set of ‘primed to death'' diffuse large B-cell lymphoma (DL-BCL) cell lines containing elevated Bcl-2 levels. We discovered a large heterogeneity in the apoptotic responses of these cells to TAT-IDPS with SU-DHL-4 being most sensitive and OCI-LY-1 being most resistant. This sensitivity strongly correlated with the ability of TAT-IDPS to promote IP3R-mediated Ca2+ release. Although total IP3R-expression levels were very similar among SU-DHL-4 and OCI-LY-1, we discovered that the IP3R2-protein level was the highest for SU-DHL-4 and the lowest for OCI-LY-1. Strikingly, TAT-IDPS-induced Ca2+ rise and apoptosis in the different DL-BCL cell lines strongly correlated with their IP3R2-protein level, but not with IP3R1-, IP3R3- or total IP3R-expression levels. Inhibiting or knocking down IP3R2 activity in SU-DHL-4-reduced TAT-IDPS-induced apoptosis, which is compatible with its ability to dissociate Bcl-2 from IP3R2 and to promote IP3-induced pro-apoptotic Ca2+ signaling. Thus, certain chronically activated B-cell lymphoma cells are addicted to high Bcl-2 levels for their survival not only to neutralize pro-apoptotic Bcl-2-family members but also to suppress IP3R hyperactivity. In particular, cancer cells expressing high levels of IP3R2 are addicted to IP3R/Bcl-2 complex formation and disruption of these complexes using peptide tools results in pro-apoptotic Ca2+ signaling and cell death.  相似文献   

9.
Smooth muscle contraction is regulated by changes in cytosolic Ca2+ concentration ([Ca2+]i). In response to stimulation, Ca2+ increase in a single cell can propagate to neighbouring cells through gap junctions, as intercellular Ca2+ waves. To investigate the mechanisms underlying Ca2+ wave propagation between smooth muscle cells, we used primary cultured rat mesenteric smooth muscle cells (pSMCs). Cells were aligned with the microcontact printing technique and a single pSMC was locally stimulated by mechanical stimulation or by microejection of KCl. Mechanical stimulation evoked two distinct Ca2+ waves: (1) a fast wave (2 mm/s) that propagated to all neighbouring cells, and (2) a slow wave (20 μm/s) that was spatially limited in propagation. KCl induced only fast Ca2+ waves of the same velocity as the mechanically induced fast waves. Inhibition of gap junctions, voltage-operated calcium channels, inositol 1,4,5-trisphosphate (IP3) and ryanodine receptors, shows that the fast wave was due to gap junction mediated membrane depolarization and subsequent Ca2+ influx through voltage-operated Ca2+ channels, whereas, the slow wave was due to Ca2+ release primarily through IP3 receptors. Altogether, these results indicate that temporally and spatially distinct mechanisms allow intercellular communication between SMCs. In intact arteries this may allow fine tuning of vessel tone.  相似文献   

10.
Although inositol trisphosphate (IP3) functions in releasing Ca2+ in eggs at fertilization, it is not known how fertilization activates the phospholipase C that produces IP3. To distinguish between a role for PLCγ, which is activated when its two src homology-2 (SH2) domains bind to an activated tyrosine kinase, and PLCβ, which is activated by a G protein, we injected starfish eggs with a PLCγ SH2 domain fusion protein that inhibits activation of PLCγ. In these eggs, Ca2+ release at fertilization was delayed, or with a high concentration of protein and a low concentration of sperm, completely inhibited. The PLCγSH2 protein is a specific inhibitor of PLCγ in the egg, since it did not inhibit PLCβ activation of Ca2+ release initiated by the serotonin 2c receptor, or activation of Ca2+ release by IP3 injection. Furthermore, injection of a PLCγ SH2 domain protein mutated at its phosphotyrosine binding site, or the SH2 domains of another protein (the phosphatase SHP2), did not inhibit Ca2+ release at fertilization. These results indicate that during fertilization of starfish eggs, activation of phospholipase Cγ by an SH2 domain-mediated process stimulates the production of IP3 that causes intracellular Ca2+ release.  相似文献   

11.

Background

In frog skeletal muscle, two ryanodine receptor (RyR) isoforms, α-RyR and β-RyR, are expressed in nearly equal amounts. However, the roles and significance of the two isoforms in excitation-contraction (E-C) coupling remains to be elucidated.

Methodology/Principal Findings

In this study, we expressed either or both α-RyR and β-RyR in 1B5 RyR-deficient myotubes using the herpes simplex virus 1 helper-free amplicon system. Immunological characterizations revealed that α-RyR and β-RyR are appropriately expressed and targeted at the junctions in 1B5 myotubes. In Ca2+ imaging studies, each isoform exhibited caffeine-induced Ca2+ transients, an indicative of Ca2+-induced Ca2+ release (CICR). However, the fashion of Ca2+ release events was fundamentally different: α-RyR mediated graded and sustained Ca2+ release observed uniformly throughout the cytoplasm, whereas β-RyR supported all-or-none type regenerative Ca2+ oscillations and waves. α-RyR but not β-RyR exhibited Ca2+ transients triggered by membrane depolarization with high [K+]o that were nifedipine-sensitive, indicating that only α-RyR mediates depolarization-induced Ca2+ release. Myotubes co-expressing α-RyR and β-RyR demonstrated high [K+]o-induced Ca2+ transients which were indistinguishable from those with myotubes expressing α-RyR alone. Furthermore, procaine did not affect the peak height of high [K+]o-induced Ca2+ transients, suggesting minor amplification of Ca2+ release by β-RyR via CICR in 1B5 myotubes.

Conclusions/Significance

These findings suggest that α-RyR and β-RyR provide distinct intracellular Ca2+ signals in a myogenic cell line. These distinct properties may also occur in frog skeletal muscle and will be important for E-C coupling.  相似文献   

12.
Insulin is released from the islets of Langerhans in discrete pulses that are linked to synchronized oscillations of intracellular free calcium ([Ca2+]i). Associated with each synchronized oscillation is a propagating calcium wave mediated by Connexin36 (Cx36) gap junctions. A computational islet model predicted that waves emerge due to heterogeneity in β-cell function throughout the islet. To test this, we applied defined patterns of glucose stimulation across the islet using a microfluidic device and measured how these perturbations affect calcium wave propagation. We further investigated how gap junction coupling regulates spatiotemporal [Ca2+]i dynamics in the face of heterogeneous glucose stimulation. Calcium waves were found to originate in regions of the islet having elevated excitability, and this heterogeneity is an intrinsic property of islet β-cells. The extent of [Ca2+]i elevation across the islet in the presence of heterogeneity is gap-junction dependent, which reveals a glucose dependence of gap junction coupling. To better describe these observations, we had to modify the computational islet model to consider the electrochemical gradient between neighboring β-cells. These results reveal how the spatiotemporal [Ca2+]i dynamics of the islet depend on β-cell heterogeneity and cell-cell coupling, and are important for understanding the regulation of coordinated insulin release across the islet.  相似文献   

13.
For decades, studies have been focusing on the neuronal abnormalities that accompany neurodegenerative disorders. Yet, glial cells are emerging as important players in numerous neurological diseases. Astrocytes, the main type of glia in the central nervous system , form extensive networks that physically and functionally connect neuronal synapses with cerebral blood vessels. Normal brain functioning strictly depends on highly specialized cellular cross-talk between these different partners to which Ca2 +, as a signaling ion, largely contributes. Altered intracellular Ca2 + levels are associated with neurodegenerative disorders and play a crucial role in the glial responses to injury. Intracellular Ca2 + increases in single astrocytes can be propagated toward neighboring cells as intercellular Ca2 + waves, thereby recruiting a larger group of cells. Intercellular Ca2+ wave propagation depends on two, parallel, connexin (Cx) channel-based mechanisms: i) the diffusion of inositol 1,4,5-trisphosphate through gap junction channels that directly connect the cytoplasm of neighboring cells, and ii) the release of paracrine messengers such as glutamate and ATP through hemichannels (‘half of a gap junction channel’). This review gives an overview of the current knowledge on Cx-mediated Ca2 + communication among astrocytes as well as between astrocytes and other brain cell types in physiology and pathology, with a focus on the processes of neurodegeneration and reactive gliosis. Research on Cx-mediated astroglial Ca2 + communication may ultimately shed light on the development of targeted therapies for neurodegenerative disorders in which astrocytes participate. This article is part of a Special Issue entitled: Calcium signaling in health and disease. Guest Editors: Geert Bultynck, Jacques Haiech, Claus W. Heizmann, Joachim Krebs, and Marc Moreau.  相似文献   

14.
Information flow within and between cells depends significantly on calcium (Ca2+) signaling dynamics. However, the biophysical mechanisms that govern emergent patterns of Ca2+ signaling dynamics at the organ level remain elusive. Recent experimental studies in developing Drosophila wing imaginal discs demonstrate the emergence of four distinct patterns of Ca2+ activity: Ca2+ spikes, intercellular Ca2+ transients, tissue-level Ca2+ waves, and a global “fluttering” state. Here, we used a combination of computational modeling and experimental approaches to identify two different populations of cells within tissues that are connected by gap junction proteins. We term these two subpopulations “initiator cells,” defined by elevated levels of Phospholipase C (PLC) activity, and “standby cells,” which exhibit baseline activity. We found that the type and strength of hormonal stimulation and extent of gap junctional communication jointly determine the predominate class of Ca2+ signaling activity. Further, single-cell Ca2+ spikes are stimulated by insulin, while intercellular Ca2+ waves depend on Gαq activity. Our computational model successfully reproduces how the dynamics of Ca2+ transients varies during organ growth. Phenotypic analysis of perturbations to Gαq and insulin signaling support an integrated model of cytoplasmic Ca2+ as a dynamic reporter of overall tissue growth. Further, we show that perturbations to Ca2+ signaling tune the final size of organs. This work provides a platform to further study how organ size regulation emerges from the crosstalk between biochemical growth signals and heterogeneous cell signaling states.  相似文献   

15.
16.
A database search of the Paramecium genome reveals 34 genes related to Ca2+-release channels of the inositol-1,4,5-trisphosphate (IP3) or ryanodine receptor type (IP3R, RyR). Phylogenetic analyses show that these Ca2+ release channels (CRCs) can be subdivided into six groups (Paramecium tetraurelia CRC-I to CRC-VI), each one with features in part reminiscent of IP3Rs and RyRs. We characterize here the P. tetraurelia CRC-IV-1 gene family, whose relationship to IP3Rs and RyRs is restricted to their C-terminal channel domain. CRC-IV-1 channels localize to cortical Ca2+ stores (alveolar sacs) and also to the endoplasmic reticulum. This is in contrast to a recently described true IP3 channel, a group II member (P. tetraurelia IP3RN-1), found associated with the contractile vacuole system. Silencing of either one of these CRCs results in reduced exocytosis of dense core vesicles (trichocysts), although for different reasons. Knockdown of P. tetraurelia IP3RN affects trichocyst biogenesis, while CRC-IV-1 channels are involved in signal transduction since silenced cells show an impaired release of Ca2+ from cortical stores in response to exocytotic stimuli. Our discovery of a range of CRCs in Paramecium indicates that protozoans already have evolved multiple ways for the use of Ca2+ as signaling molecule.Ca2+ is an important component of cell activity in all organisms, from protozoa to mammals. Thereby Ca2+ may originate from the outside medium and/or from internal stores (7, 18). Ca2+ release from internal stores is mediated by various Ca2+ release channels (CRCs), of which the inositol-1,4,5-trisphosphate receptor (IP3R) and ryanodine receptor (RyR) families have been studied most extensively (8, 9, 29, 63). IP3Rs and RyRs have been identified in various metazoan organisms (reviewed in references 9, 28, and 104). According to these reviews, there exist three genetically distinct isoforms of each receptor type in mammals and orthologues have been identified in various nonmammalian vertebrates, e.g., frogs, chickens, and fish. RyRs and IP3Rs were also cloned and sequenced in the invertebrates Drosophila melanogaster and Caenorhabditis elegans, which possess one copy of each receptor type.Functional evidence for Ca2+ release in response to ryanodine or IP3 receptor agonists has been described in several unicellular systems. Treatment of permeabilized Plasmodium chabaudi parasites with IP3 results in Ca2+ release, which is inhibited by the IP3 receptor antagonist heparin (69). Another apicomplexan parasite, Toxoplasma gondii, responds to agonists and antagonists of both, ryanodine and IP3 receptors, by mediating increases in intracellular Ca2+ concentration ([Ca2+]i) (56). Stimulation of Trypanosoma cruzi with carbachol results in increased [Ca2+]i and IP3 (59). IP3 and cyclic ADP-ribose induces Ca2+ release in Euglena gracilis microsome fractions in a dose-dependent manner (61). In the giant algae Chara corallina and Nitrella translucens, IP3 produces action potentials involving increased [Ca2+]i (93). Treatment of vacuolar membrane vesicles from Candida albicans with IP3 results in Ca2+ release, blocked by heparin and ruthenium red (14). IP3 generates and maintains a Ca2+ gradient in the hyphal tip of Neurospora crassa and the IP3-sensitive channels have been reconstituted and characterized with the planar bilayer method (87). In summary, these publications suggest that IP3-dependent signaling pathways are conserved among unicellular organisms, including protozoa.Despite these data, the molecular characterization of IP3 or ryanodine receptors in low eukaryotes is currently a challenge since the identification of orthologues has not been possible thus far, probably because of evolutionary sequence divergence (66). Traynor et al. (96) identified an IP3 receptor-like protein, IplA, in Dictyostelium discoideum, which possesses regions related to IP3R sequences, but thus far no evidence for IP3 interaction exists. We have recently described an IP3R in the ciliated protozoa Paramecium tetraurelia (referred to here as P. tetraurelia IP3RN) (53), with features characteristic of mammalian IP3Rs in terms of topology and ability for IP3 binding. The expression level of P. tetraurelia IP3RN is modulated by extracellular Ca2+ concentrations ([Ca2+]o) and immunofluorescence studies reveal an unexpected localization to the contractile vacuole complex (CVC), the major organelle involved in osmoregulation (2). The ionic composition of the contractile vacuole fluid by ion-selective microelectrodes (91) suggests that the organelle plays a major role in expelling an excess of cytosolic Ca2+. Therefore, these IP3Rs may here mediate a latent, graded reflux of Ca2+ for fine-tuning of [Ca2+]i and thus serve [Ca2+] homeostasis (53).Besides [Ca2+] homeostasis, the Paramecium cell has to regulate a variety of well-characterized processes (75). This includes exocytosis of dense-core secretory vesicles (trichocysts) (71, 74, 99). Each cell possesses up to 1,000 trichocysts attached to the cell membrane. Their contents can be extruded synchronously in response to natural stimuli, i.e., predators (34, as confirmed by Knoll et al. [49]), to artificial polyamine secretagogues such as aminoethyldextran (AED) (78), to caffeine (48) or to the ryanodine substitute, 4-chloro-meta-cresol (4-CmC) (46). Their expulsion strictly depends on Ca2+ (10) and is accompanied by an increase of intracellular [Ca2+]i (24, 47). This Ca2+ signal originates from rapid mobilization of cortical stores, the alveolar sacs (33, 64, 74), superimposed by Ca2+ influx (46, 72). It thus represents a SOC-type mechanism (SOC, store-operated Ca2+ entry) known from mammalian systems (81).Upon exocytosis stimulation ∼60% of their total Ca2+ is released from alveolar sacs (33). These are Ca2+ stores (90) represented by flat membrane compartments tightly attached at the cell membrane surrounding each trichocyst docking site. They possess a SERCA-type pump located at the membrane facing the cell center (36, 37) and a luminal high-capacity/low-affinity CaBP of the calsequestrin type (73). Thus far, Ca2+ release channels of these stores were identified only indirectly as cells respond by exocytosis to the RyR activators caffeine (54, 48) and 4-CmC (46). However, an involvement of conserved RyRs has remained questionable as ryanodine is not able to activate Ca2+ release from alveolar sacs, as is the case with IP3 (54). Therefore, one of the most intriguing questions is the elucidation of the molecular nature of the channels mediating Ca2+ release from alveolar sacs upon stimulated exocytosis.In the present work we describe a novel family of CRCs (P. tetraurelia CRC-IV-1), whose members display several properties of the channels postulated above. In detail, the identified CRC-IV-1 channels localize to the alveolar sacs. Functional and fluorochrome analyses after gene silencing reveal that they are essential for mediating Ca2+ release and exocytosis in response to AED, caffeine, or 4-CmC. Their classification as “novel” CRC type is based on a restricted relationship to the C-terminal channel domains of IP3Rs and RyRs. The overall size and the number of putative transmembrane domains resemble IP3Rs, but N-terminal parts of CRC-IV-1 channels do not show any conservation, such as an IP3-binding domain. Therefore, CRC-IV-1 channels represent distant relatives of IP3Rs and RyRs and may belong to an ancestral Ca2+ signaling pathway.  相似文献   

17.

Background

Abnormal Ca2+ transients are often observed in heart muscles under a variety of pathophysiological conditions including ventricular tachycardia. To clarify whether these abnormal Ca2+ transients can be attributed to abnormal action potential generation or abnormal Ca2+ handling/excitation-contraction (EC) coupling, we developed a procedure to determine Ca2+ and action potential signals at the cellular level in isolated heart tissues.

Methodology/Principal Findings

After loading ventricular papillary muscle with rhod-2 and di-4-ANEPPS, mono-wavelength fluorescence images from rhod-2 and ratiometric images of two wavelengths of emission from di-4-ANEPPS were sequentially obtained. To mimic the ventricular tachycardia, the ventricular muscles were field-stimulated in non-flowing Krebs solution which elicited abnormal Ca2+ transients. For the failed and alternating Ca2+ transient generation, there were two types of causes, i.e., failed or abnormal action potential generation and abnormal EC coupling. In cells showing delayed initiation of Ca2+ transients with field stimulation, action potential onset was delayed and the rate of rise was slower than in healthy cells. Similar delayed onset was also observed in the presence of heptanol, an inhibitor of gap junction channels but having a non-specific channel blocking effect. A Na+ channel blocker, on the other hand, reduced the rate of rise of the action potentials but did not result in desynchronization of the action potentials. The delayed onset of action potentials can be explained primarily by impaired gap junctions and partly by Na+ channel inactivation.

Conclusions/Significance

Our results indicate that there are multiple patterns for the causes of abnormal Ca2+ signals and that our methods are useful for investigating the physiology and pathophysiology of heart muscle.  相似文献   

18.
Collet C  Ma J 《Biophysical journal》2004,87(1):268-275
Activation of store-operated Ca2+ entry (SOCE) into the cytoplasm requires retrograde signaling from the intracellular Ca2+ release machinery, a process that involves an intimate interaction between protein components on the intracellular and cell surface membranes. The cellular machinery that governs the Ca2+ movement in muscle cells is developmentally regulated, reflecting maturation of the junctional membrane structure as well as coordinated expression of related Ca2+ signaling molecules. Here we demonstrate the existence of SOCE in freshly isolated skeletal muscle cells obtained from embryonic days 15 and 16 of the mouse embryo, a critical stage of muscle development. SOCE in the fetal muscle deactivates incrementally with the uptake of Ca2+ into the sarcoplasmic reticulum (SR). A novel Ca2+-dependent facilitation of SOCE is observed in cells transiently exposed to high cytosolic Ca2+. Our data suggest that cytosolic Ca2+ can facilitate SOCE whereas SR luminal Ca2+ can deactivate SOCE in the fetal skeletal muscle. This cooperative mechanism of SOCE regulation by Ca2+ ions not only enables tight control of SOCE by the SR membrane, but also provides an efficient mechanism of extracellular Ca2+ entry in response to physiological demand. Such Ca2+ signaling mechanism would likely contribute to contraction and development of the fetal skeletal muscle.  相似文献   

19.
S Finkbeiner 《Neuron》1992,8(6):1101-1108
Stimulus-evoked cellular responses are sometimes organized in the form of propagating waves of cytoplasmic Ca2+ increase. Ca2+ waves can be elicited in cultured astrocytes by the neurotransmitter glutamate; however, the propagation mechanism is unknown. Here, qualitative and quantitative features of propagation suggest that astrocytic Ca2+ waves are mediated by an intracellular signal that crosses intercellular junctions. The role of gap junctions in cell-cell Ca2+ wave propagation was specifically tested. Functional gap junctions were demonstrated using a noninvasive fluorescence recovery method and the gap junction blockers halothane and octanol. Gap junction closure prevented intracellular waves from propagating between cells without affecting the velocity of the intracellular wave itself. The pivotal role played by the gap junction creates the potential for dynamic changes in glial connectivity and long-range glial signaling.  相似文献   

20.
Calcium deficiency causes abnormal colonic growth and increases colon cancer risk with poorly understood mechanisms. Here we elucidate a novel signaling mechanism underlying the Ca2+ deficiency-induced epithelial proliferation using a unique animal model. The zebrafish larval yolk sac skin contains a group of Ca2+-transporting epithelial cells known as ionocytes. Their number and density increases dramatically when acclimated to low [Ca2+] environments. BrdU pulse-labeling experiments suggest that low [Ca2+] stimulates pre-existing ionocytes to re-enter the cell cycle. Low [Ca2+] treatment results in a robust and sustained activation of IGF1R-PI3K-Akt signaling in these cells exclusively. These ionocytes specifically express Igfbp5a, a high-affinity and specific binding protein for insulin-like growth factors (IGFs) and the Ca2+-selective channel Trpv5/6. Inhibition or knockdown of Igfbp5a, IGF1 receptor, PI3K, and Akt attenuates low [Ca2+]-induced ionocyte proliferation. The role of Trpv5/6 was investigated using a genetic mutant, targeted knockdown, and pharmacological inhibition. Loss-of-Trpv5/6 function or expression results in elevated pAkt levels and increased ionocyte proliferation under normal [Ca2+]. These increases are eliminated in the presence of an IGF1R inhibitor, suggesting that Trpv5/6 represses IGF1R-PI3K-Akt signaling under normal [Ca2+]. Intriguingly, blockade of Trpv5/6 activity inhibits the low [Ca2+]-induced activation of Akt. Mechanistic analyses reveal that the low [Ca2+]-induced IGF signaling is mediated through Trpv5/6-associated membrane depolarization. Low extracellular [Ca2+] results in a similar amplification of IGF-induced PI3K-PDK1-Akt signaling in human colon cancer cells in a TRPV6-dependent manner. These results uncover a novel and evolutionarily conserved signaling mechanism that contributes to the abnormal epithelial proliferation associated with Ca2+ deficiency.  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