Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Basic Res Cardiol ; 108(1): 309, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23184389

ABSTRACT

Connexin-43 (Cx43), a predominant cardiac connexin, forms gap junctions (GJs) that facilitate electrical cell-cell coupling and unapposed/nonjunctional hemichannels that provide a pathway for the exchange of ions and metabolites between cytoplasm and extracellular milieu. Uncontrolled opening of hemichannels in the plasma membrane may be deleterious for the myocardium and blocking hemichannels may confer cardioprotection by preventing ionic imbalance, cell swelling and loss of critical metabolites. Currently, all known hemichannel inhibitors also block GJ channels, thereby disturbing electrical cell-cell communication. Here we aimed to characterize a nonapeptide, called Gap19, derived from the cytoplasmic loop (CL) of Cx43 as a hemichannel blocker and examined its effect on hemichannel currents in cardiomyocytes and its influence in cardiac outcome after ischemia/reperfusion. We report that Gap 19 inhibits Cx43 hemichannels without blocking GJ channels or Cx40/pannexin-1 hemichannels. Hemichannel inhibition is due to the binding of Gap19 to the C-terminus (CT) thereby preventing intramolecular CT-CL interactions. The peptide inhibited Cx43 hemichannel unitary currents in both HeLa cells exogenously expressing Cx43 and acutely isolated pig ventricular cardiomyocytes. Treatment with Gap19 prevented metabolic inhibition-enhanced hemichannel openings, protected cardiomyocytes against volume overload and cell death following ischemia/reperfusion in vitro and modestly decreased the infarct size after myocardial ischemia/reperfusion in mice in vivo. We conclude that preventing Cx43 hemichannel opening with Gap19 confers limited protective effects against myocardial ischemia/reperfusion injury.


Subject(s)
Connexin 43/antagonists & inhibitors , Ion Channels/drug effects , Myocardial Reperfusion Injury/prevention & control , Peptide Fragments/pharmacology , Adenosine Triphosphate/metabolism , Animals , Gap Junctions/drug effects , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Swine
2.
PLoS One ; 7(7): e42074, 2012.
Article in English | MEDLINE | ID: mdl-22860057

ABSTRACT

ATP-dependent paracrine signaling, mediated via the release of ATP through plasma membrane-embedded hemichannels of the connexin family, coordinates a synchronized response between neighboring cells. Connexin 43 (Cx43) hemichannels that are present in the plasma membrane need to be tightly regulated to ensure cell viability. In monolayers of bovine corneal endothelial cells (BCEC),Cx43-mediated ATP release is strongly inhibited when the cells are treated with inflammatory mediators, in particular thrombin and histamine. In this study we investigated the involvement of RhoA activation in the inhibition of hemichannel-mediated ATP release in BCEC. We found that RhoA activation occurs rapidly and transiently upon thrombin treatment of BCEC. The RhoA activity correlated with the onset of actomyosin contractility that is involved in the inhibition of Cx43 hemichannels. RhoA activation and inhibition of Cx43-hemichannel activity were both prevented by pre-treatment of the cells with C3-toxin as well as knock down of RhoA by siRNA. These findings provide evidence that RhoA activation is a key player in thrombin-induced inhibition of Cx43-hemichannel activity. This study demonstrates that RhoA GTPase activity is involved in the acute inhibition of ATP-dependent paracrine signaling, mediated by Cx43 hemichannels, in response to the inflammatory mediator thrombin. Therefore, RhoA appears to be an important molecular switch that controls Cx43 hemichannel openings and hemichannel-mediated ATP-dependent paracrine intercellular communication under (patho)physiological conditions of stress.


Subject(s)
Connexin 43/physiology , rhoA GTP-Binding Protein/physiology , Adenosine Triphosphate/metabolism , Animals , Cattle , Cells, Cultured , Connexin 43/genetics , Endothelium/cytology , Endothelium/metabolism , Endothelium/physiology , Gene Knockdown Techniques , RNA, Small Interfering , rhoA GTP-Binding Protein/genetics
3.
FASEB J ; 26(9): 3649-57, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22665389

ABSTRACT

Recent in vitro evidence indicates that astrocytes can modulate synaptic plasticity by releasing neuroactive substances (gliotransmitters). However, whether gliotransmitter release from astrocytes is necessary for higher brain function in vivo, particularly for memory, as well as the contribution of connexin (Cx) hemichannels to gliotransmitter release, remain elusive. Here, we microinfused into the rat basolateral amygdala (BLA) TAT-Cx43L2, a peptide that selectively inhibits Cx43-hemichannel opening while maintaining synaptic transmission or interastrocyte gap junctional communication. In vivo blockade of Cx43 hemichannels during memory consolidation induced amnesia for auditory fear conditioning, as assessed 24 h after training, without affecting short-term memory, locomotion, or shock reactivity. The amnesic effect was transitory, specific for memory consolidation, and was confirmed after microinfusion of Gap27, another Cx43-hemichannel blocker. Learning capacity was recovered after coinfusion of TAT-Cx43L2 and a mixture of putative gliotransmitters (glutamate, glutamine, lactate, d-serine, glycine, and ATP). We propose that gliotransmitter release from astrocytes through Cx43 hemichannels is necessary for fear memory consolidation at the BLA. Thus, the present study is the first to demonstrate a physiological role for astroglial Cx43 hemichannels in brain function, making these channels a novel pharmacological target for the treatment of psychiatric disorders, including post-traumatic stress disorder.


Subject(s)
Amygdala/physiology , Astrocytes/metabolism , Connexin 43/metabolism , Fear , Memory , Neurotransmitter Agents/metabolism , Animals , Cells, Cultured , Connexin 43/antagonists & inhibitors , Immunohistochemistry , Rats , Rats, Sprague-Dawley
4.
Biol Cell ; 104(7): 367-77, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22375941

ABSTRACT

The molecular mechanisms underlying the regulation of gap junction (GJ) channels based on the 43-kDa connexin isoform (Cx43) have been studied extensively. GJ channels are formed by the docking of opposed hemichannels in adjacent cells. Mounting data indicate that unopposed Cx43 hemichannels are also functional in the plasma membrane. However, our understanding of how Cx43-hemichannel opening and closing is regulated at the molecular level is only poorly understood. Recent work elucidated that actomyosin contractility inhibits potently Cx43 hemichannels. It is known that intracellular Ca²âº exerts a bell-shaped-dependent effect on Cx43-hemichannel opening. While low-intracellular [Ca²âº] (<500 nM) provokes opening of the channel, high-intracellular [Ca²âº] (> 500 nM) favours closing of the channel. The mechanism underlying this negative regulation of Cx43-hemichannel activity by high-intracellular [Ca²âº] seems to be dependent on the activation of the actomyosin contractile system. The activity of Cx43 hemichannels is critically controlled by molecular interactions between the intracellular loop and the C-terminal tail. These interactions are essential for Cx43-hemichannel opening in response to triggers such as cytosolic [Ca²âº] rise or external [Ca²âº] lowering. In this review, we present the hypothesis that the actomyosin contractile system can function as an important brake mechanism on Cx43-hemichannel opening. By controlling loop-tail interactions, the contractile system would prevent aberrant or excessive opening of Cx43 hemichannels.


Subject(s)
Actomyosin/metabolism , Connexin 43/metabolism , Gap Junctions/metabolism , Animals , Cells/metabolism , Connexin 43/chemistry , Humans
5.
J Biol Chem ; 287(15): 12250-66, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22351781

ABSTRACT

Many cellular functions are driven by changes in the intracellular Ca(2+) concentration ([Ca(2+)](i)) that are highly organized in time and space. Ca(2+) oscillations are particularly important in this respect and are based on positive and negative [Ca(2+)](i) feedback on inositol 1,4,5-trisphosphate receptors (InsP(3)Rs). Connexin hemichannels are Ca(2+)-permeable plasma membrane channels that are also controlled by [Ca(2+)](i). We aimed to investigate how hemichannels may contribute to Ca(2+) oscillations. Madin-Darby canine kidney cells expressing connexin-32 (Cx32) and Cx43 were exposed to bradykinin (BK) or ATP to induce Ca(2+) oscillations. BK-induced oscillations were rapidly (minutes) and reversibly inhibited by the connexin-mimetic peptides (32)Gap27/(43)Gap26, whereas ATP-induced oscillations were unaffected. Furthermore, these peptides inhibited the BK-triggered release of calcein, a hemichannel-permeable dye. BK-induced oscillations, but not those induced by ATP, were dependent on extracellular Ca(2+). Alleviating the negative feedback of [Ca(2+)](i) on InsP(3)Rs using cytochrome c inhibited BK- and ATP-induced oscillations. Cx32 and Cx43 hemichannels are activated by <500 nm [Ca(2+)](i) but inhibited by higher concentrations and CT9 peptide (last 9 amino acids of the Cx43 C terminus) removes this high [Ca(2+)](i) inhibition. Unlike interfering with the bell-shaped dependence of InsP(3)Rs to [Ca(2+)](i), CT9 peptide prevented BK-induced oscillations but not those triggered by ATP. Collectively, these data indicate that connexin hemichannels contribute to BK-induced oscillations by allowing Ca(2+) entry during the rising phase of the Ca(2+) spikes and by providing an OFF mechanism during the falling phase of the spikes. Hemichannels were not sufficient to ignite oscillations by themselves; however, their contribution was crucial as hemichannel inhibition stopped the oscillations.


Subject(s)
Calcium Signaling , Connexin 43/metabolism , Cytoplasm/metabolism , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Bradykinin/pharmacology , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Carbenoxolone/pharmacology , Cell Line , Connexin 43/genetics , Connexins/metabolism , Cytochromes c/metabolism , Cytochromes c/physiology , Dogs , Fluoresceins/metabolism , Gene Knockdown Techniques , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Oligopeptides/pharmacology , Peptides/pharmacology , RNA Interference , Rats , Recombinant Proteins/metabolism , Gap Junction beta-1 Protein
6.
Autophagy ; 7(12): 1472-89, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22082873

ABSTRACT

The role of intracellular Ca2+ signaling in starvation-induced autophagy remains unclear. Here, we examined Ca2+ dynamics during starvation-induced autophagy and the underlying molecular mechanisms. Tightly correlating with autophagy stimulation, we observed a remodeling of the Ca2+ signalosome. First, short periods of starvation (1 to 3 h) caused a prominent increase of the ER Ca2+-store content and enhanced agonist-induced Ca2+ release. The mechanism involved the upregulation of intralumenal ER Ca2+-binding proteins, calreticulin and Grp78/BiP, which increased the ER Ca2+-buffering capacity and reduced the ER Ca2+ leak. Second, starvation led to Ins(1,4,5)P3R sensitization. Immunoprecipitation experiments showed that during starvation Beclin 1, released from Bcl-2, first bound with increasing efficiency to Ins(1,4,5)P3Rs; after reaching a maximal binding after 3 h, binding, however, decreased again. The interaction site of Beclin 1 was determined to be present in the N-terminal Ins(1,4,5)P3-binding domain of the Ins(1,4,5)P3R. The starvation-induced Ins(1,4,5)P3R sensitization was abolished in cells treated with BECN1 siRNA, but not with ATG5 siRNA, pointing toward an essential role of Beclin 1 in this process. Moreover, recombinant Beclin 1 sensitized Ins(1,4,5)P3Rs in 45Ca2+-flux assays, indicating a direct regulation of Ins(1,4,5)P3R activity by Beclin 1. Finally, we found that Ins(1,4,5)P3R-mediated Ca2+ signaling was critical for starvation-induced autophagy stimulation, since the Ca2+ chelator BAPTA-AM as well as the Ins(1,4,5)P3R inhibitor xestospongin B abolished the increase in LC3 lipidation and GFP-LC3-puncta formation. Hence, our results indicate a tight and essential interrelation between intracellular Ca2+ signaling and autophagy stimulation as a proximal event in response to starvation.


Subject(s)
Autophagy , Calcium Signaling , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 5 , Beclin-1 , Binding Sites , Calcium/metabolism , Calcium-Binding Proteins/metabolism , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Gene Knockdown Techniques , HeLa Cells , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate Receptors/chemistry , Intracellular Space/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Models, Biological , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/metabolism
7.
Biochim Biophys Acta ; 1815(1): 13-25, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20801193

ABSTRACT

It is nowadays well established that gap junctions are critical gatekeepers of cell proliferation, by controlling the intercellular exchange of essential growth regulators. In recent years, however, it has become clear that the picture is not as simple as originally anticipated, as structural precursors of gap junctions can affect cell cycling by performing actions not related to gap junctional intercellular communication. Indeed, connexin hemichannels also foresee a pathway for cell growth communication, albeit between the intracellular compartment and the extracellular environment, while connexin proteins as such can directly or indirectly influence the production of cell cycle regulators independently of their channel activities. Furthermore, a novel set of connexin-like proteins, the pannexins, have lately joined in as regulators of the cell proliferation process, which they can affect as either single units or as channel entities. In the current paper, these multifaceted aspects of connexin-related signalling in cell cycling are reviewed.


Subject(s)
Cell Cycle , Connexins/physiology , Animals , Cell Proliferation , Connexins/chemistry , Connexins/genetics , Gene Expression Regulation , Humans , Signal Transduction
8.
Cell Signal ; 23(2): 305-16, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20688156

ABSTRACT

The pannexin (Panx) family of proteins, which is co-expressed with connexins (Cxs) in vertebrates, was found to be a new GJ-forming protein family related to invertebrate innexins. During the past ten years, different studies showed that Panxs mainly form hemichannels in the plasma membrane and mediate paracrine signalling by providing a flux pathway for ions such as Ca²(+), for ATP and perhaps for other compounds, in response to physiological and pathological stimuli. Although the physiological role of Panxs as a hemichannel was questioned, there is increasing evidence that Panx play a role in vasodilatation, initiation of inflammatory responses, ischemic death of neurons, epilepsy and in tumor suppression. Moreover, it is intriguing that Panxs may also function at the endoplasmic reticulum (ER) as intracellular Ca²(+)-leak channel and may be involved in ER-related functions. Although the physiological significance and meaning of such Panx-regulated intracellular Ca²(+) leak requires further exploration, this functional property places Panx at the centre of many physiological and pathophysiological processes, given the fundamental role of intracellular Ca²(+) homeostasis and dynamics in a plethora of physiological processes. In this review, we therefore want to focus on Panx as channels at the plasma membrane and at the ER membranes with a particular emphasis on the potential implications of the latter in intracellular Ca²(+) signalling.


Subject(s)
Adenosine Triphosphate/metabolism , Connexins/physiology , Endoplasmic Reticulum/metabolism , Animals , Calcium Signaling/physiology , Cell Membrane/metabolism , Humans , Mice
9.
FASEB J ; 24(11): 4378-95, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20634352

ABSTRACT

Connexin-assembled gap junctions (GJs) and hemichannels coordinate intercellular signaling processes. Although the regulation of connexins in GJs has been well characterized, the molecular determinants controlling connexin-hemichannel activity are unresolved. Here we investigated the regulation of Cx43-hemichannel activity by actomyosin contractility and intracellular [Ca(2+)] ([Ca(2+)](i)) using plasma membrane-permeable TAT peptides (100 µM) designed to interfere with interactions between the cytoplasmic loop (CL) and carboxy-terminal (CT) in primary bovine corneal endothelial cells and HeLa, C6 glioma, and Xenopus oocytes ectopically expressing Cx43. Peptides corresponding to the last 10 CT aa (TAT-Cx43CT) prevented the inhibition of Cx43-hemichannel activity by contractility/high [Ca(2+)](i), whereas a reverse peptide (TAT-Cx43CTrev) did not. These effects were independent of zonula occludens-1, a cytoskeletal-associated Cx43-binding protein. In contrast, peptides corresponding to CL (TAT-L2) inhibited Cx43-hemichannel responses, whereas a mutant peptide (TAT-L2(H126K/I130N)) did not inhibit. In these assays, TAT-Cx43CT acted as a scaffold for TAT-L2 and vice versa, a finding supported by surface plasmon resonance measurements. Loop/tail interactions appeared essential for Cx43-hemichannel activity, because TAT-Cx43CT restored the activity of nonfunctional hemichannels, consisting of either Cx43 lacking the C-terminal tail (Cx43(M239)) or intact Cx43 ectopically expressed in Xenopus oocytes. We conclude that intramolecular loop/tail interactions control Cx43-hemichannel activity, laying the basis for developing hemichannel-specific blockers.


Subject(s)
Connexin 43/metabolism , Ion Channels/metabolism , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Cattle , Cell Line , Cell Line, Tumor , Cornea/cytology , Cornea/metabolism , Endothelial Cells/metabolism , Gene Products, tat/metabolism , HeLa Cells , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Intracellular Space/metabolism , Ion Channels/drug effects , Oocytes/metabolism , Protein Binding , Rats , Thrombin/metabolism , Xenopus laevis/metabolism
10.
Curr Eye Res ; 34(6): 454-65, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19899980

ABSTRACT

PURPOSE: Mechanical stimulation induces intercellular Ca(2 +) waves in the corneal endothelium. The extent of the wave propagation is dependent on the activity of gap junctions, hemichannels, and ectonucleotidases. To further establish the use of a cell culture model to investigate intercellular communication, in this study, we have characterized the changes in the Ca(2 +) wave propagation in bovine corneal endothelial cells with prolonged time in culture. MATERIALS AND METHODS: Freshly isolated BCEC were cultured for a short term (8 to 14 days; referred to as "short term") and a long term (21 to 30 days; referred to as "long term"). Cell surface area and size were measured by confocal microscopy and flow cytometry, respectively. Calcium wave propagation was assayed by imaging spread of the Ca(2 +) waves elicited by mechanical stimulation. ATP release was assayed using Luciferin-Luciferase bioluminescence technique. RESULTS: Cells cultured for a long term showed larger surface area and size compared to those cultured for a short term, but a reduced spread of the Ca(2 +) wave. Exposure to exogenous apyrases, which can rapidly hydrolyze extracellular ATP, reduced the spread of the Ca(2 +) wave in both groups. The fractional decrease, however, was smaller in cells cultured for a long term. Exposure to ARL-67156 to inhibit the ectonucleotidases led to a larger enhancement of the active area in cells cultured for a long term. However, the active areas of the two groups were not significantly different in the presence of the drug. Furthermore, ATP release in response to mechanical stimulation was lower in cells cultured for a long term in the absence of ARL-67156 but not in its presence. CONCLUSIONS: BCEC cultured for a long term show an increase in cell surface area and cell size similar to the effect of aging in human corneas. Moreover, the cells cultured for a long term showed a reduced ATP-dependent paracrine intercellular communication, largely due to an increase in the activity of the ectonucleotidases.


Subject(s)
Cell Communication/physiology , Endothelium, Corneal/cytology , Endothelium, Corneal/physiology , Actins , Adenosine Triphosphatases/metabolism , Animals , Calcium/metabolism , Cattle , Cell Culture Techniques , Cell Size , Flow Cytometry , Time Factors , Tubulin
11.
Bioessays ; 31(9): 953-74, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19644918

ABSTRACT

Intercellular communication (IC) is mediated by gap junctions (GJs) and hemichannels, which consist of proteins. This has been particularly well documented for the connexin (Cx) family. Initially, Cxs were thought to be the only proteins capable of GJ formation in vertebrates. About 10 years ago, however, a new GJ-forming protein family related to invertebrate innexins (Inxs) was discovered in vertebrates, and named the pannexin (Panx) family. Panxs, which are structurally similar to Cxs, but evolutionarily distinct, have been shown to be co-expressed with Cxs in vertebrates. Both protein families show distinct properties and have their own particular function. Identification of the mechanisms that control Panx channel gating is a major challenge for future work. In this review, we focus on the specific properties and role of Panxs in normal and pathological conditions.


Subject(s)
Connexins/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Communication , Connexins/chemistry , Humans , Intercellular Junctions/chemistry , Intercellular Junctions/metabolism , Ion Channel Gating , Protein Transport
12.
Invest Ophthalmol Vis Sci ; 49(11): 4816-27, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18614806

ABSTRACT

PURPOSE: Thrombin inhibits intercellular Ca(2+) wave propagation in bovine corneal endothelial cells (BCECs) through a mechanism dependent on myosin light chain (MLC) phosphorylation. In this study, blebbistatin, a selective myosin II ATPase inhibitor, was used to investigate whether the effect of thrombin is mediated by enhanced actomyosin contractility. METHODS: BCECs were exposed to thrombin (2 U/mL) for 5 minutes. MLC phosphorylation was assayed by immunocytochemistry. Ca(2+) waves were visualized by confocal microscopy with Fluo-4AM. Fluorescence recovery after photobleaching (FRAP) was used to investigate intercellular communication (IC) via gap junctions. ATP release was measured by luciferin-luciferase assay. Lucifer yellow (LY) uptake was used to investigate hemichannel activity, and Fura-2 was used to assay thrombin- and ATP-mediated Ca(2+) responses. RESULTS: Pretreatment with blebbistatin (5 microM for 20 minutes) or its nitro derivative prevented the thrombin-induced inhibition of the Ca(2+) wave. Neither photo-inactivated blebbistatin nor the inactive enantiomers prevented the thrombin effect. Blebbistatin also prevented thrombin-induced inhibition of LY uptake, ATP release and FRAP, indicating that it prevented the thrombin effect on paracrine and gap junctional IC. In the absence of thrombin, blebbistatin had no significant effect on paracrine or gap junctional IC. The drug had no influence on MLC phosphorylation or on [Ca(2+)](i) transients in response to thrombin or ATP. CONCLUSIONS: Blebbistatin prevents the inhibitory effects of thrombin on intercellular Ca(2+) wave propagation. The findings demonstrate that myosin II-mediated actomyosin contractility plays a central role in thrombin-induced inhibition of gap junctional IC and of hemichannel-mediated paracrine IC.


Subject(s)
Calcium Channels/drug effects , Calcium/metabolism , Endothelium, Corneal/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Intracellular Fluid/metabolism , Thrombin/pharmacology , Adenosine Triphosphatases/antagonists & inhibitors , Animals , Blotting, Western , Calcium Channels/metabolism , Cattle , Endothelium, Corneal/cytology , Endothelium, Corneal/drug effects , Fluorescent Dyes/pharmacokinetics , Gap Junctions/drug effects , Gap Junctions/metabolism , Gene Expression , Hemostatics/pharmacology , Immunohistochemistry , Isoquinolines/pharmacokinetics , Myosin Type II/antagonists & inhibitors , Myosin Type II/biosynthesis , Myosin Type II/genetics , Phosphorylation/drug effects , Physical Stimulation , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction
13.
Invest Ophthalmol Vis Sci ; 48(1): 120-33, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17197525

ABSTRACT

PURPOSE: Thrombin, a serine protease, breaks down the barrier integrity of corneal endothelial cells by phosphorylation of the regulatory light chain of myosin II (myosin light chain; MLC), which induces contractility of the actin cytoskeleton. This study was undertaken to investigate the effect of thrombin on gap junctional (GJIC) and paracrine (PIC) intercellular communication in cultured bovine corneal endothelial cells (BCECs). METHODS: An intercellular Ca(2+) wave, a form of cell-cell communication, was elicited by applying a mechanical stimulus to a single cell in a confluent monolayer. Changes in [Ca(2+)](i) were imaged by fluorescence microscopy with a fluorescent calcium indicator, and the images were used to calculate the area reached by the Ca(2+) wave (active area). GJIC was assessed by fluorescence recovery after photobleaching (FRAP). Activity of hemichannels was assayed by lucifer yellow (LY) uptake and also by adenosine triphosphate (ATP) release by using the luciferin-luciferase technique. RESULTS: RT-PCR showed transcripts for PAR-1 and -2 receptors, but not for PAR-4 receptors. Immunocytochemistry showed thrombin-sensitive PAR receptors as well as trypsin-sensitive PAR-2 receptors. Both thrombin and the selective PAR-1 agonist TRAP-6 reduced the active area of the Ca(2+) wave. These agents also reduced the fluorescence recovery in FRAP experiments. The effect of thrombin on the Ca(2+) wave was inhibited by a peptide antagonist of PAR-1, but not by a PAR-4 antagonist. Pretreatment with ML-7 (an MLCK inhibitor), Y-27632 (a Rho kinase inhibitor) or chelerythrine (a PKC inhibitor) prevented the effect of thrombin on the Ca(2+) wave. Activation of PAR-1 did not affect the Ca(2+) wave propagation in cells pretreated with Gap26, which blocks hemichannels. However, PAR-1 activation decreased the active area in cells pretreated with Gap27, which inhibits gap junctions. Thrombin abolished enhancement of the Ca(2+) wave propagation by ARL-67156 (inhibitor of ecto-ATPases). The effect of the PAR-1 agonists on the Ca(2+) wave was not detectable in cells pretreated with exogenous apyrases. CONCLUSIONS: Thrombin inhibits intercellular Ca(2+) wave propagation in BCECs. This effect is due to activation of PAR-1 receptors and involves MLC phosphorylation by MLCK-, PKC- and Rho kinase-sensitive pathways. Thrombin mainly inhibits the ATP-mediated PIC pathway, and also reduces GJIC to a lesser extent.


Subject(s)
Calcium/metabolism , Endothelium, Corneal/drug effects , Gap Junctions/drug effects , Paracrine Communication/drug effects , Receptor, PAR-1/metabolism , Receptor, PAR-2/metabolism , Thrombin/pharmacology , Adenosine Triphosphate/metabolism , Animals , Calcium Signaling/physiology , Cattle , Cells, Cultured , Endothelium, Corneal/cytology , Endothelium, Corneal/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Myosin Light Chains/metabolism , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...