Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Feb 5.
Published in final edited form as: Glia. 2008 Jan 15;56(2):145–153. doi: 10.1002/glia.20598

Modulation of Astrocyte P2Y1 Receptors by the Carboxyl Terminal Domain of the Gap Junction Protein Cx43

Eliana Scemes 1
PMCID: PMC2636557  NIHMSID: NIHMS79211  PMID: 17990308

Abstract

Gap junction proteins, connexins, provide intercellular channels that allow ions and small signaling molecules to be transmitted to adjacent coupled cells. Besides this function, it is becoming apparent that connexins also exert channel-independent effects, which are likely mediated by processes involving protein–protein interactions. Although a number of connexin interacting proteins have been identified, only little is known about the functional consequences of such interactions. We have previously shown that deletion of the astrocytic gap junction protein, connexin43 (Cx43) causes a right-ward shift in the dose-response curve to P2Y1R agonists and decreased P2Y1R expression levels. To evaluate whether these changes were due to reduced gap junctional communication or to protein–protein interactions, Cx43-null astrocytes were transfected with full-length Cx43 and Cx43 domains, and P2Y1R function and expression levels evaluated. Results indicate that restoration of P2Y1R function is independent of gap junctional communication and that the Cx43 carboxyl terminus spanning the SH3 binding domain (260–280) participates in the rescue of P2Y1R pharmacological behavior (shifting to the left the P2Y1R dose-response curve) without affecting its expression levels. These results suggest that the Cx43 carboxyl-terminus domain provides a binding site for an intracellular molecule, most likely a member of the c-Src tyrosine kinase family, which affects P2Y1R-induced calcium mobilization. It is here proposed that a nonchannel function of Cx43 is to serve as a decoy for such kinases. Such modulation of P2Y1R is expected to influence several neural cell functions, especially under inflammation and neurodegenerative disorders where expression levels of Cx43 are decreased.

Keywords: glia, calcium signaling, connexin, purinergic receptor

INTRODUCTION

The gap junction protein connexin43 (Cx43) is widely expressed in most tissues (Beyer et al., 1987; Willecke et al., 2002), including the central nervous system (CNS). During early stages of CNS development, Cx43 expression is highly abundant in both neuronal and glial cell lineages (Duval et al., 2002; Rozental et al., 1998, 2000) but decreases as the cells differentiate and mature into neurons and oligodendrocytes, being restricted to the astrocytic glial cell population (Dermietzel et al., 1991; Rash et al., 2001a,b). Cx43 forms intercellular channels permeable to ions and molecules up to 900 Da. By providing continuity and exchange of signals between the interconnected intracellular compartments, Cx43 gap junction channels play important roles in astrocyte biology as well as during CNS development. Several factors can either decrease or increase intercellular communication between cells. For instance, closure of Cx43 gap junction channels in astrocytes and in other cell types may result from intracellular acidification (Dermietzel et al., 1991; Duffy et al., 2004; Ek-Vitorin et al., 1996; Liu et al., 1993; for review see Spray and Scemes, 1998), or as a consequence of changes in phosphorylation state following activation of protein kinases (Cooper et al., 2000; Kwak and Jongsma, 1996; Lampe et al., 2000; Li et al., 2005; Saez et al., 1998), whereas junctional conductance is increased by high extracellular K+ (Enkvist and McCarthy, 1994; Pina-Benabou et al., 2001), and membrane permeant cAMP derivatives (Burghardt et al., 1995; Chanson et al., 1996; Darrow et al., 1996; Matesic et al., 1996; Paulson et al., 2000).

Recently, it has become apparent that connexins, specifically Cx43, may have other functions that are independent of intercellular channel formation. For instance it has been shown that forced expression of Cx43 in C6 glioma cells provides resistance to apoptotic signals (Lin et al., 2003a) and that over-expression or deletion of the Cx43 gene alters the expression of numerous unrelated gene products (Iacobas et al., 2003, 2004; Naus et al., 2000), including certain membrane receptors (Suadicani et al., 2003). One possible way by which connexins may exert channel independent effects may be through the action of Cx43 interacting proteins with either transcriptional activity, such as β-catenin (Ai et al., 2000; Xu et al., 2001) and members of src family of oncogenes (Kanemitsu et al., 1997; Loo et al., 1995), or with others with structural functions, such as the scaffolding protein ZO1 (Giepmans and Moolenaar, 1998; Toyofuku et al., 1998), and the microtubule protein tubulin (Giepmans et al., 2001), as well with the lipid raft protein caveolin-1 (Lin et al., 2003b; Schubert et al., 2002). Such functional diversity of molecules that have been shown to colocalize and to coimmunoprecipitate with Cx43, strongly suggest that Cx43 is a component of several supramolecular complexes (Duffy et al., 2002a; Spray et al., 1999). Among the cytoplasmic domains of Cx43, the carboxyl-terminus (CT) contains SH3, SH2 and PDZ binding sites that have been shown to be important for interaction with these proteins (for review see Delmar et al., 2004; Giepmans, 2004). Although the list of gap junction interacting proteins continues to grow, very little is known about the functional significance of such interactions. Our previous work on spinal cord astrocytes indicated that deletion of Cx43 alters the expression and function of purinergic (P2Y) receptors, thus affecting glial calcium signal transmission (Scemes et al., 2000b, Suadicani et al., 2003) and the rate of migration of neural progenitor cells (Scemes et al., 2003). Such effects of Cx43 on P2YR expression/function led us to hypothesize that these two proteins are part of a functional unit involving protein–protein interactions. To test this possibility, P2Y1R function was evaluated in Cx43-null astrocytes transfected with full length Cx43, Cx43 truncated at amino acid residue 257, the Cx43 carboxyl terminus alone, and with a Cx43 construct lacking residues 260–280. Evidence is here provided for the functional interaction of Cx43 and P2Y1R in astrocytes, involving the SH3 binding domain on the carboxyl terminus of Cx43. Such change in P2Y1 receptor function is likely to have great impact in the CNS, especially under inflammatory conditions due to the decreased expression of Cx43.

MATERIALS AND METHODS

Astrocyte Cultures

Spinal cord astrocytes were obtained as previously described (Scemes et al., 2000b) from wild-type (WT) and Cx43-null neonatal mice (C57Bl/6J-Gja1 strain, heterozygotes were originally obtained from Jackson Laboratories and colonies maintained at the Albert Einstein College of Medicine; all experimental procedures were approved by the AECOM Animal Care and Use Committee). Briefly, cervical to lumbar vertebrae were dissected and spinal cord segments evicted from the vertebrae. After removal of the meninges, spinal cord tissues were digested in 0.025% collagenase (Sigma) for 10 min at 37°C. Cells were collected by centrifugation at 2,000 rpm for 2 min at room temperature. The final pellet was suspended in Dulbecco’s Modified Eagle Medium (DMEM, Gibco) containing 5% fetal bovine serum and 1% antibiotics and cells seeded in 35 mm culture dishes. Studies were performed on spinal cord astrocytes maintained for 2–3 weeks in culture at which time about 95–98% of the cells in culture were immunopositive for glial fibrillary acidic protein.

Transient Transfection of Cx4-Null Astrocytes with Cx43 cDNAs

Confluent cultures of Cx43-null spinal cord astrocytes were transfected with 6–8 µg of cDNA encoding either full length Cx43, Cx43 truncated at position 257 (M257), with Cx43 CT (255–382) or with Cx43Δ260–280 using lipofectamine 2000 (Invitrogen). Parallel experiments were performed on mock-treated (lipofectamine and/or empty vector) Cx43-null and WT spinal cord astrocytes. Cells were used 30–48 h after transfection. Cx43 constructs were originally obtained from Dr. Eric Beyer (full length Cx43) and Dr. Mario Delmar (Cx43M257, Cx43Δ260–280 and Cx43CT).

Western Blots

Samples of whole cell lysates of WT and of untransfected and transfected Cx43-null astrocytes were electrophoresed in 4–20% SDS-PAGE (Bio-Rad, Hercules, CA) and then transferred to nitrocellulose membranes (Schleicher & Schuell, Keene, NH). Immunoblots were performed after overnight incubation of membranes with blocking solution (5% dry nonfat milk in 1X PBS) using primary antibodies. After several washes with 1X PBS containing 0.4% polyoxyethylenesorbitan monolaurate (Tween-20; Sigma), membranes were incubated with HRP-conjugated secondary antibodies (1:2000; Santa Cruz Biotechnology, Santa Cruz, CA). Detection of bands was performed on X-ray films (Kodak, Rochester, NY) following incubation with enhanced chemiluminescence reagents (Amersham Pharmacia Biotechnology, Piscataway, NJ). Quantification of the expression level of Cx43 and P2Y1R was performed by densitometric analysis (Scion-NIH Image software) using β-actin as an invariant control against which the protein of interest was normalized. The following primary antibodies were employed: polyclonal antiP2Y1 (1:200; Alomone Labs), and two different polyclonal antiCx43 antibodies (gifts from Dr. E.L. Hertzberg, Albert Einstein College of Medicine), one recognizing Cx43 at position 241–261 (16A) and the other at position 345–360 (18A).

Intracellular Calcium Transients

Agonist-induced intracellular Ca2+ transients were measured in WT and in mock-treated and transfected Cx43-null astrocytes loaded for 45 min at 37°C with Fura-2-AM (10 µM; Molecular Probes, as previously described (Scemes et al., 2000b; Suadicani et al., 2003). Cells were then washed with Dulbecco’s phosphate buffered saline (DPBS; pH 7.4; Cellgro, Herndon, VA) containing 5 mM glucose and imaged on an epifluorescence microscope (Eclipse TE2000-S; Nikon, Japan). Intracellular calcium measurements were performed on cells plated on Matek dishes and bathed in DPBS. Images were acquired with a CCD camera (Orca-ER; Hamamatsu, Japan). Ratio values of Fura-2 fluorescence intensities emitted at two excitation wavelengths (340 and 380 nm) were obtained using combined systems of filters and shutter (Lambda DG-4 Diaphot, Sutter Instruments Co., Burlingame, CA) driven by a computer through Metafluor software (Universal Imaging Systems, PA) and acquired continuously at a rate of 0.7 Hz. Measurements were obtained from regions of interest placed on cells, and ratio values were translated into intracellular Ca2+ concentrations according to an in vitro calibration curve of the form: [Ca2+]i=KD{(RRmin)/(RmaxR)}(Fmin380/Fmax380), where [Ca2+]i is the calculated intracellular calcium concentration, KD is the dissociation constant of free-Ca2+ for Fura-2 (KD = 224 nM; Grynkiewicz et al., 1985), R is the ratio intensity, Rmin is the ratio of the intensity obtained at zero calcium, Rmax is the ratio of the intensity at saturated calcium, Fmin380 is the fluorescence intensity measured with zero calcium at 380 nm, and Fmax380 is the fluorescence intensity measured with saturated calcium at 380 nm. Changes in intracellular calcium levels were monitored before and after bath application of the P2Y1R agonist 2-methylthioadenosine 5′-triphosphate (2-MeS-ATP; Calbiochem, La Jolla, CA), as previously described (Scemes et al., 2000b; Suadicani et al., 2003).

For some experiments aimed to evaluate the responses of P2Y1R to agonist, Cx43-null astrocytes were treated for 30 min with 10 µM of a membrane permeant (TAT) peptide-FITC conjugated (GenScript) corresponding to amino acids 260–280 of Cx43CT and with the c-Src tyrosine kinase inhibitor 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4,d]pyrimidine (PP2;10 µM; Tocris).

Fluorescence Recovery After Photobleach

Evaluation of dye-coupling in WT, Cx43-null and Cx43-null astrocytes transiently transfected with full length Cx43 and truncated Cx43Δ257 was performed using the fluorescence recovery after photobleaching (FRAP) technique ( Lippincott-Schwartz et al., 2001; Wade et al., 1986), as previously described (Suadicani et al., 2004). Confluent cultures of astrocytes plated in Matek dishes were loaded with calcein-AM (1 µM; Molecular Probes) for 30 min at 37°C. After several washes, cells were bathed in DPBS (Dulbecco’s phosphate buffered salt solution; pH 7.4, Cellgro) throughout the experiments performed at RT. Changes in calcein fluorescence intensity emitted at 510 nm when excited at 488 nm were monitored over time using a real time confocal microscope (Nikon RCM 8000) equipped with a 40× water immersion objective. Photobleach of calcein was attained within seconds by directing the laser bean, set at maximum power, to one to two cells using a 4× optical zoom (inserted between the light path and the objective). Immediately after the photobleach, changes in calcein fluorescence intensity were then monitored at low laser power and 1× optical zoom. Images were acquired continuously at a rate of 1/60 s before, immediately after and for about 15 min following photobleach. OMDR stored images were played back and calcein fluorescence intensity values measured from regions of interest placed on the bleached cells using Polygon-Star software (Nikon, Japan). Evaluation of the degree of dye coupling was obtained by measuring the half times of fluorescence recovery, as follows. The absolute values of calcein fluorescence intensity obtained after photobleach (t = 1–15 min) were subtracted from the ones obtained immediately after photobleach (t = 0) and then normalized to the values obtained at the plateau phase of fluorescence recovery (t = 15 min). The half-time (t1/2) of FRAP was obtained using GraphPad Prism4.0 curve fitting software according to the following exponential equation: Y = Ymax [1 − exp(−Kt)]; where Y is the change in fluorescence intensity over time, Ymax is the maximum fluorescence intensity attained at t = 15 min after photobleach and K is the rate constant (t1/2 = 0.69/K). The t1/2 values were then used as parameters to evaluate the degree of dye-coupling in WT and Cx43-null astrocytes. At least three independent experiments were performed.

RESULTS

To evaluate whether changes in P2Y1R expression and function observed in Cx43-null spinal cord astrocytes (Scemes et al., 2000b; Suadicani et al., 2003) were related to reduced coupling between Cx43-null cells or mediated through specific domains of the Cx43 protein, experiments described below were performed on Cx43-null spinal cord astrocytes transiently transfected with vectors encoding full length Cx43, Cx43 truncated at position 257 (M257), and the Cx43 carboxyl terminus (Cx43CT: amino acids 255–382). Parallel experiments were performed on WT astrocytes treated with the gap junction channel blocker carbenoxolone (CBX: 100 µM).

Expression of Cx43 Domains and Dye Coupling in Astrocytes

As illustrated in Western blots in Fig. 1A, transient transfection of Cx43-null astrocytes with full length Cx43, M257 and Cx43CT resulted in readily detectable expression of these three proteins. Functional studies performed using the FRAP assay (Fig. 1B) indicated that expression of both full length Cx43 and M257 provided dye coupling between Cx43-null astrocytes. The half-time of calcein recovery after photobleaching obtained for cells transfected with full length Cx43 (85.4 ± 2.91 s; N = 15 cells from three to four independent experiments) was not statistically different (P > 0.05, t-test) from that observed between WT cells (91.3 ± 0.72 s; N = 15 cells). Interestingly, dye coupling between cells expressing Cx43M257, which in oocytes produces pH-insensitive gap junction channels (Morley et al., 1996), was significantly lower (111.2 ± 0.28 s; N = 15 cells; P < 0.001, t-test) than WT cells. The possibility that such difference in dye coupling is related to distinct expression levels of each of the connexin constructs used cannot be ruled out. Because the two Cx43 antibodies used (18A and 16A) for western blot analyses are likely to have different affinities for the protein, comparison of band intensities generated by these two antibodies is unlikely to correlate with Cx43 expression levels.

Fig. 1. Gap junctional communication.

Fig. 1

(A) Western blot showing expression of Cx43 in WT (lane 1) and in Cx43-null astrocytes transfected with full length Cx43 (lane 3), with Cx43 carboxyl terminus (lane 4) and with Cx43M257 (lane 5). Lane 2 corresponds to untransfected Cx43-null cells. Lanes 1–4 correspond to immunoblots performed with Cx43-18A antibody and lane 5 with Cx43-16A. (B) Time courses of calcein FRAP obtained from untreated (black squares) and 48 h carbenoxolone-treated (open triangles) WT astrocytes, and from untransfected (open circles) and transfected Cx43-null astrocytes with full length Cx43 (open squares) and Cx43M257 (black circles). Data were obtained from astrocytes cultured from a minimum of 3 litters of mice.

Treatment of WT astrocytes with 100 µM CBX for 48 h (incubation time chosen to correspond to that evaluated after transfection) reduced dye coupling to levels similar to those seen in Cx43-null astrocytes (Fig. 1B); no changes in Cx43 expression levels were observed after CBX treatment (data not shown).

Expression of Specific Cx43 Domain But Not Blockade of Coupling Affects P2Y1R Function

To evaluate whether the altered P2Y1R function in Cx43-null astrocytes (Scemes et al., 2000b) was related to reduced gap junctional communication in these cells, experiments were performed by measuring the half-maximal increases in intracellular Ca2+ (EC50 values) induced by the P2Y1R agonist 2-MeS-ATP in Fura-2-AM-loaded WT astrocytes untreated and treated with the gap junction channel blocker CBX. Figure 2A shows the dose-response curves obtained from untreated and CBX-treated WT and Cx43-null astrocytes. Exposure of WT spinal cord astrocytes to 100 µM CBX for 48 h did not significantly alter the EC50 value to 2-MeS-ATP (EC50 = 11.8 ± 0.7 nM; N = 8 independent experiments) compared with those obtained for WT cells (EC50 = 12.9 ± 0.7 nM; N = 8 experiments), which were significantly different from those obtained for Cx43-null astrocytes (EC50 = 380.5 ± 20.0 nM; N = 8 experiments). Note that CBX treatment totally uncoupled astrocytes, as shown in Fig. 1. These data, therefore, indicate that altered P2Y1R function in Cx43-null spinal cord astrocytes is unlikely to be due to reduced gap junctional communication between these cells and suggest that other mechanisms contribute to the change in agonist EC50 values.

Fig. 2. Modulation of P2Y1R activity by Cx43 domains.

Fig. 2

(A) Dose-response curves obtained for untreated (black squares) and 48 h carbenoxolone-treated (open triangles) WT and untransfected Cx43-null spinal cord astrocytes exposed to the P2Y1R agonist 2-MeS-ATP. Note that blockade of gap junctional communication in WT astrocytes does not alter the half-maximal response (EC50 value) induced by the P2Y1R agonist. Mean values were obtained from four to seven independent experiments. (B) Dose-response curves obtained for WT (black squares), untransfected (open circles) and transfected Cx43-null astrocytes with full length Cx43 (open squares), Cx43M257 (black circles) and Cx43CT (black triangles). Note that both full length Cx43 and Cx43CT but not Cx43M257 shifted the EC50 values of 2-MeS-ATP obtained for Cx43-null astrocytes to those obtained in WT cells. Data were obtained from 5 to 8 litters.

That the effect of high concentrations of 2-MeSATP (10 and 100 µM) used in the present study is not mediated by P2X receptors relies on the observation that calcium transients induced by 10 and 100 µM MeSATP were totally prevented by the specific P2Y1R antagonist MRS2179 (5 µM) (see Fig. 4 in Scemes et al., 2003; Fig. S1 in supplemental material). Moreover, bath application of 300 µM α,β-meATP, an agonist of P2X1 and P2X3 receptors, which are sensitive to 2-MeSATP (Burnstock, 2007), failed to induce calcium elevation in astrocytes (Fig. S1 in supplemental material), as previously reported (Fumagalli et al., 2003; Scemes et al., 2000a).

Fig. 4. P2Y1R expression levels in Cx43-null transfectants.

Fig. 4

Bar histograms showing the mean values of P2Y1R expression levels in WT, untransfected Cx43-null (KO), and in Cx43-null transfected with Cx43 CT, Cx43 truncated at position 257 (M257) and with full length Cx43. An example of western blots for Cx43 and β-actin is shown above.

To test the hypothesis that P2Y1R function is dependent on a mechanism that requires the expression of Cx43, Cx43-null spinal cord astrocytes were transiently transfected with full length Cx43 and the half maximal responses to 2-MeS-ATP were measured in Fura-2-AM loaded cells, as described above. As shown in Fig. 2B, expression of full length Cx43 in Cx43-null astrocytes rescued P2Y1R behavior; the half-maximal value of intracellular Ca2+ increase induced by the P2Y1R agonist observed in mock-treated Cx43-null astrocytes (EC50 = 321.0 ± 24.6 nM; N = 12 experiments) shifted to 42.1 ± 6.6 nM (N = 9 experiments) in full length Cx43 transfectants; the EC50 values obtained for transfected Cx43-null cells were not different (P > 0.05; ANOVA) from those obtained for WT cells (EC50 = 38.5 ± 4.9 nM; N = 10 experiments). Thus, these data strongly suggest that expression of Cx43 protein is important for the proper function of P2Y1R.

To evaluate whether specific Cx43 domains mediated the rescue of P2Y1R function, dose-response curves to 2-MeS-ATP were performed in Cx43-null astrocytes transfected with Cx43CT and with M257. As shown in Fig. 2B, forced expression of Cx43CT in Cx43-null astrocytes, but not of M257, shifted the dose-response curve obtained for 2-MeS-ATP toward that obtained for WT cells. The EC50 values obtained for 2-MeS-ATP in Cx43CT transfectants (52.7 ± 1.6 nM; N = 10 experiments) were similar to those obtained for WT cells and full length Cx43 transfectants (P > 0.05; ANOVA). No significant difference (P > 0.05; ANOVA) was observed between M257 transfectants (EC50 = 254.1 ± 30.2 nM; N = 10 experiments) and mock-treated Cx43-null cells (EC50 = 321.0 ± 24.6 nM; N = 12 experiments).

The domain of Cx43CT spanning 264–287 comprises an SH3 binding site, which has been shown to play an important role in the competition between c-Src and ZO1 (Sorgen et al., 2004). Therefore, we hypothesized that the effect of Cx43CT on P2Y1R pharmacology could be mediated by a member of the Src tyrosine kinase family. To test this hypothesis, Cx43-null astrocytes were transfected with a Cx43 mutant construct missing the SH3 domain (deletion of amino acids 260–280). As shown in Fig. 3A, transfection of Cx43-null astrocytes with Cx43Δ260–280 did not rescue the EC50 value of 2-MeS-ATP observed in WT cells (EC50 = 849.6 nM for Cx43Δ260–280; EC50 = 40.0 nM for WT). Conversely, treatment of Cx43-null astrocytes with a membrane permeant peptide corresponding to amino acids 260–280 of Cx43CT shifted the dose-response curve to the left (Fig. 3B; EC50 = 49.4 nM and 57.2 nM for peptide and EC50 = 16.4 nM for WT).

Fig. 3. Functional interaction between P2Y1R and the SH3 domain of Cx43.

Fig. 3

(A) Dose-response curves obtained for WT (black squares) and untransfected (open circles) and Cx43Δ260–280 transfected (black triangles) Cx43-null astrocytes exposed to 2-MeS-ATP, showing that deletion of the Cx43 SH3 domain does not rescue P2Y1 receptor function. Mean±SE values are from 100 to 200 cells obtained from three litters. (B) Dose-response curves obtained from WT (black squares), and Cx43-null astrocytes untreated (open circles) and treated (black triangles) with a membrane permeant peptide corresponding to amino acids 260–280 of Cx43CT. (C) Bar histograms of the mean±SE values of intracellular calcium mobilization induced by 100 nM 2-MeS-ATP recorded from WT, Cx43-null and Cx43-null transfected with Cx43CT and Cx43M257 in the absence and presence of 5 µM PP2. Note that only untransfected and Cx43M257 transfected Cx43-null astrocytes that were not exposed to PP2 did not respond to agonist with intracellular calcium levels similar to WT astrocytes. Mean ± SE values are from 4 litters (**P < 0.001; ANOVA followed by Newman-Keuls’ Multiple Comparison Test).

These results suggest that altered P2Y1R function in Cx43-null astrocytes is dependent on the expression of Cx43, specifically the Cx43 carboxyl terminal spanning the SH3 domain of the c-Src binding site. To further test this hypothesis, WT, untransfected and transfected Cx43-null astrocytes were treated with the c-Src kinase inhibitor PP2 (10 µM) prior to stimulation with 2-MeS-ATP (100 nM). As shown in Fig. 3C, intracellular calcium mobilization induced by 2-MeS-ATP was enhanced in Cx43-null astrocytes treated with PP2 to levels similar to those recorded from WT and from Cx43-null astrocytes transfected with Cx43CT; intracellular calcium mobilization was not altered in astrocytes expressing the truncation mutant Cx43M257 compared with untransfected cells.

P2Y1R Expression Levels in Cx43-Null Transfectants

To evaluate whether the rescue of P2Y1R function in Cx43-null transfectants was related to increased expression levels of P2Y1R in these cells, western blot analyses were performed in untransfected and transfected Cx43-null astrocytes with the three different Cx43 constructs. As shown in Fig. 4, the reduced expression level of P2Y1R in Cx43-null astrocytes was not restored to that measured from WT cells following forced expression of either Cx43CT or M257 (P < 0.001; ANOVA followed by Newman-Keuls Multiple Comparison Test). Interestingly, although the levels of P2Y1R in full length Cx43 transfectants were not statistically different from that of WT, they also did not significantly differ from that seen in Cx43-null cells (P > 0.05; ANOVA followed by Newman-Keuls Multiple Comparison Test).

These results showing that expression of Cx43 domains in Cx43-null astrocytes do not restore P2Y1R expression levels suggest that the increased agonist sensitivity of P2Y1R induced by forced expression of full length Cx43 and Cx43CT (Fig. 2B) is independent of either direct or indirect dependence of transcriptional activity of Cx43 on P2Y1R expression levels. These data, however, raise the possibility that the binding of Cx43 to a tyrosine kinase of the Src family facilitates calcium mobilization induced by P2Y1R activation.

Altered P2Y1 Receptor Function Following Cytokine-Induced Decrease in Cx43

Given that Cx43 expression levels in astrocytes can be greatly reduced under inflammatory conditions (Brand-Schieber et al., 2005; Hinkerohe et al., 2005; John et al., 1999; Meme et al., 2006), we evaluated whether such decrease could also affect P2Y1R function. For that, we used confluent cultures of WT spinal cord astrocytes treated with the proinflammatory cytokine interleukin (IL)-1β to evaluate Cx43 expression levels and P2Y1R induced calcium mobilization. Western blot analysis of whole cell homogenates of confluent cultures of untreated and IL1-β treated (20 ng/mL; Cedarlene) WT mouse spinal cord astrocytes showed, as expected, a significant decrease of Cx43 expression levels 6 h after treatment (Fig. 5) that was further decreased after 12 and 24 h. The dose-response curves obtained for the P2Y1R agonist 2-MeS-ATP performed on WT treated for 24 h with IL-1β yield EC50 values of 291.5 ± 1.3 nM (see Fig. 5), which are about 10–20 times higher than those observed for untreated WT astrocytes (see Fig. 2 and Fig. 3). Because no significant changes in the P2Y1R agonist EC50 values were observed between untreated and IL-1β treated Cx43-null astrocytes (Fig. 2, Fig. 3, and Fig. 5), it is likely that the altered P2Y1R behavior observed under these conditions is related to the reduced expression of Cx43 rather than to downstream effects of IL-1β on P2Y1R. Moreover, no significant changes in P2Y1R expression levels were observed in astrocytes treated for 24 h with the cytokine (Fig. S2 in supplemental material). Similarly to what was here observed for IL-1β, activation of Toll-like receptor 3 in human fetal astrocytes, which also leads to decreased Cx43 expression level, does not alter P2Y1R receptor expression levels (Zhao et al., 2006).

Fig. 5. Effect of IL-1β on Cx43 and P2Y1R.

Fig. 5

(A) Western blot showing decreased expression levels of Cx43 following exposure of spinal cord astrocytes to IL-1β (B) Dose-response curves obtained for 2-MeS-ATP performed on Fura-2 loaded WT and Cx43 KO astrocytes treated for 24 h with IL-1β (20 ng/mL). Note that exposure to the cytokine altered the agonist EC50 values in WT astrocytes. About 180 cells from three independent experiments were used in each condition.

DISCUSSION

Astrocytes “in vivo” and “in vitro” are equipped with distinct purinergic P2 receptors (Ballerini et al., 1996; Fumagalli et al., 2003; Jimenez et al., 2000; Neary et al., 1988; Pearce and Langley, 1994; Suadicani et al., 2003; Zhu and Kimelberg, 2001) and gap junction proteins (Dermietzel et al., 2000; Nagy et al., 2003; Scemes et al., 2000b; Spray et al., 1998). Although these protein families are genetically unrelated, they share at least the common function of regulating calcium levels in astrocytes. Activation of P2 receptors by exogenous purine and pyrimidine nucleotides leads to elevation of intracellular calcium levels that can be constrained within a single cell or transmitted through gap junction channels to adjacent coupled astrocytes (for reviews see Scemes, 2000; Scemes and Giaume, 2006). Changes in cytosolic calcium levels are not only important for several aspects of astrocyte biology but can also impact on neuronal activity due to the exocytotic release of ATP and glutamate from astrocytes (Bezzi et al., 2004; Montana et al., 2004; Striedinger et al., 2007; Zhang et al., 2004). Because the main gap junction protein in astrocytes (Cx43) forms intercellular channels permeable to Ca2+ mobilizing second messengers, such as IP3, cADP-ribose, Ca2+ (Christ et al., 1992; Churchill and Louis, 1998; Saez et al., 1989), Cx43 gap junctional channels are expected to modulate P2R mediated Ca2+ signals within the astrocytic compartment. Indeed, we have recently shown that gap junctional communication between glial cells in culture can either increase or decrease the number of cells responding to P2R activation depending on the levels of second messenger generated by each P2Y receptor subtype (Suadicani et al., 2004).

It is becoming apparent that gap junction proteins have roles other than providing direct ionic and metabolic continuity between the cytosol of coupled cells. Previous studies performed on Cx43-null astrocytes indicated that numerous unrelated genes are altered, such as those involved in cell cycle, motility and metabolism (Iacobas et al., 2003, 2004; Naus et al., 2000). The mechanisms by which connexins exert channel- independent effects are not clear. It is possible that they are mediated by connexin interacting proteins (for reviews see Delmar et al., 2004; Giepmans, 2004), implying that connexins are part of a supra-molecular complex (Duffy et al., 2002a; Spray et al., 1999).

In this regard, evidence is here provided suggesting that the major gap junction protein in astrocytes, Cx43, modulates purinergic receptor activity, altering P2Y1R agonist concentration necessary to attain half maximal intracellular calcium elevation. In accordance with our previous work showing that deletion of Cx43 by homologous recombination or by the use of anti-sense oligonucleotide altered the behavior of P2Y1R (Scemes et al., 2000b; Suadicani et al., 2003) are the results obtained here showing that IL-1β-induced decrease in Cx43 expression levels causes a similar shift in 2-MeS-ATP EC50 value as that seen in Cx43 KO astrocytes.

Moreover, it is shown here that this modulation is independent of gap junctional communication but is dependent on the presence of the Cx43 carboxyl-terminal domain. Evidence in support for a gap junction-independent modulation of P2Y1R activity includes the findings that the dose-response curves obtained for P2Y1R agonist were not affected by (1) blockade of gap junctional communication in WT astrocytes and by (2) coupling Cx43 KO astrocytes with Cx43M257 gap junction channels. In favor of P2Y1R modulation through Cx43 specific domains are the results showing that P2Y1R function is rescued when Cx43-null astrocytes were transfected with either full length Cx43 or with its CT domain (Cx43CT).

A possible mechanism by which such interplay between connexins and purinergic receptors occurs may include protein–protein interactions. For instance, it has been shown that specific sequences of the C-termini of P2Y1 and P2Y2 receptors, distinct from their PDZ-binding motif, modulate the voltage dependence of the endogenous Xenopus oocyte transient inward (Tin) channels (Lee et al., 2003). Moreover, the binding of the SH3 domain of c-Src to P2Y2R was shown to lead to the recruitment of the epidermal growth factor receptor (EGFR) into the P2Y2R complex, thus facilitating EGFR phosphorylation by c-Src (Liu et al., 2004). Although the number of Cx43 binding partners has been increasing (see Giepman, 2004; Iacobas et al., 2007), little is known about their functional consequences. Binding of the Cx43 C-terminus to its cytoplasmic loop has been proposed as a mechanism for the pH gating of Cx43 gap junction channels (Duffy et al., 2002b; Ek-Vitorin et al., 1996; Morley et al., 1996), while the binding of c-Src SH3 domain to Cx43CT, following prolonged intracellular acidification was shown to lead to the unbinding of the PDZ domain of Cx43 from the scaffolding protein ZO-1 and to Cx43 internalization (Duffy et al., 2004).

We here show that the amino acid sequence 260–280 of Cx43 CT that contains an SH3 sequence motif (Sorgen et al., 2004) is necessary for proper function of P2Y1R. This is based on the observation that a peptide spanning the amino acids 260–280 of Cx43, but not expression of Cx43 lacking this motif, was able to restore P2Y1R function. A possible mechanism by which Cx43 modulates P2Y1R is by providing a binding site for c-Src, i.e., by serving as a decoy. In such a model, reduction of a c-Src binding site (i.e., Cx43) could lead to increased binding and phosphorylation of other proteins (P2Y1R), thus altering their function. Support for a role of c-Src on P2Y1R is our observation that the Src tyrosine kinase inhibitor PP2 restored P2Y1R-induced calcium mobilization in Cx43-null astrocytes.

Given that P2Y1R plays important roles in multiple biological functions (production and release of NO from astrocytes: Horiuchi et al., 2003; astrogliosis: Franke et al., 2001; progenitor cell migration: Scemes et al., 2003; Weisman et al., 2004), the modulation of P2Y1R activity by Cx43 is likely to impact on CNS physiology and pathology, especially under conditions of CNS inflammation and in neuro-degenerative disorders, where Cx43 has been shown to be down-regulated (Brand-Schieber et al., 2005; Hinkerohe et al., 2005; John et al., 1999; Meme et al., 2006).

Supplementary Material

Figure 1S

This article contains supplementary material available via the Internet at http://www.interscience.wiley.com/jpages/0894-1491/suppmat.

Figure 2S
Figure Legends

ACKNOWLEDGMENTS

All suggestions and discussion provided by Dr. David C. Spray were greatly appreciated.

Grant sponsor: NINDS-NIH; Grant number: NS-41023.

REFERENCES

  1. Ai Z, Fischer A, Spray DC, Brown AM, Fishman GI. Wnt-1 regulation of connexin43 in cardiac myocytes. J Clin Invest. 2000;105:161–171. doi: 10.1172/JCI7798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Ballerini P, Rathbone MP, Di Iorio P, Renzetti A, Giuliani P, D’Alimonte I, Trubiani O, Caciagli F, Ciccarelli R. Rat astroglial P2Z (P2X7) receptors regulate intracellular calcium and purine release. Neuroreport. 1996;7:2533–2537. doi: 10.1097/00001756-199611040-00026. [DOI] [PubMed] [Google Scholar]
  3. Beyer EC, Paul DL, Goodenough DA. Connexin43: A protein from rat heart homologous to a gap junction protein from liver. J Cell Biol. 1987;105:2621–2629. doi: 10.1083/jcb.105.6.2621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Bezzi P, Gundersen V, Galbete JL, Seifert G, Steinhauser C, Pilati E, Volterra A. Astrocytes contain a vesicular compartment that is competent for regulated exocytosis of glutamate. Nat Neurosci. 2004;7:613–620. doi: 10.1038/nn1246. [DOI] [PubMed] [Google Scholar]
  5. Brand-Schieber E, Werner P, Iacobas DI, Iacobas S, Beelitz M, Lowery SL, Spray DC, Scemes E. Connexin43, the major gap junction protein of astrocytes, is down regulated in inflamed white matter in an animal model of multiple sclerosis. J Neurosci Res. 2005;80:798–808. doi: 10.1002/jnr.20474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Burghardt RC, Barhoumi R, Sewall TC, Bowen JA. Cyclic AMP induces rapid increases in gap junction permeability and changes in the cellular distribution of connexin43. J Membr Biol. 1995;148:243–253. doi: 10.1007/BF00235042. [DOI] [PubMed] [Google Scholar]
  7. Burnstock G. Purine and pyrimidine receptors. Cell Mol Life Sci. 2007;64:1471–1483. doi: 10.1007/s00018-007-6497-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Chanson M, White MM, Garber SS. cAMP promotes gap junctional coupling in T84 cells. Am J Physiol. 1996;271:C533–C539. doi: 10.1152/ajpcell.1996.271.2.C533. [DOI] [PubMed] [Google Scholar]
  9. Christ GJ, Moreno AP, Melman A, Spray DC. Gap junction-mediated intercellular diffusion of Ca2+ in cultured human corporal smooth muscle cells. Am J Physiol. 1992;263:C373–C383. doi: 10.1152/ajpcell.1992.263.2.C373. [DOI] [PubMed] [Google Scholar]
  10. Churchill GC, Louis CF. Roles of Ca2+, inositol trisphosphate and cyclic ADP-ribose in mediating intercellular Ca2+ signaling in sheep lens cells. J Cell Sci. 1998;111:1217–1225. doi: 10.1242/jcs.111.9.1217. [DOI] [PubMed] [Google Scholar]
  11. Cooper CD, Solan JL, Dolejsi MK, Lampe PD. Analysis of connexin phosphorylation sites. Methods. 2000;20:196–204. doi: 10.1006/meth.1999.0937. [DOI] [PubMed] [Google Scholar]
  12. Darrow BJ, Fast VG, Kleber AG, Beyer EC, Saffitz JE. Functional and structural assessment of intercellular communication. Increased conduction velocity and enhanced connexin expression in dibutyryl cAMP-treated cultured cardiac myocytes. Circ Res. 1996;79:174–183. doi: 10.1161/01.res.79.2.174. [DOI] [PubMed] [Google Scholar]
  13. Delmar M, Coombs W, Sorgen P, Duffy HS, Taffet SM. Structural bases for the chemical regulation of connexin43 channels. Cardiovasc Res. 2004;62:268–275. doi: 10.1016/j.cardiores.2003.12.030. [DOI] [PubMed] [Google Scholar]
  14. Dermietzel R, Gao Y, Scemes E, Vieira D, Urban M, Kremer M, Bennett MV, Spray DC. Connexin43 null mice reveal that astrocytes express multiple connexins. Brain Res Brain Res Rev. 2000;32:45–56. doi: 10.1016/s0165-0173(99)00067-3. [DOI] [PubMed] [Google Scholar]
  15. Dermietzel R, Hertberg EL, Kessler JA, Spray DC. Gap junctions between cultured astrocytes: Immunocytochemical, molecular, and electrophysiological analysis. J Neurosci. 1991;11:1421–1432. doi: 10.1523/JNEUROSCI.11-05-01421.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Duffy HS, Ashton AW, O’Donnell P, Coombs W, Taffet SM, Delmar M, Spray DC. Regulation of connexin43 protein complexes by intracellular acidification. Circ Res. 2004;94:215–222. doi: 10.1161/01.RES.0000113924.06926.11. [DOI] [PubMed] [Google Scholar]
  17. Duffy HS, Delmar M, Spray DC. Formation of the gap junction nexus: Binding partners for connexins. J Physiol Paris. 2002a;96:243–249. doi: 10.1016/s0928-4257(02)00012-8. [DOI] [PubMed] [Google Scholar]
  18. Duffy HS, Sorgen PL, Girvin ME, O’Donnell P, Coombs W, Taffet SM, Delmar M, Spray DC. pH-dependent intramolecular binding and structure involving Cx43 cytoplasmic domains. J Biol Chem. 2002b;277:36706–36714. doi: 10.1074/jbc.M207016200. [DOI] [PubMed] [Google Scholar]
  19. Duval N, Gomes D, Calaora V, Calabrese A, Meda P, Bruzzone R. Cell coupling and Cx43 expression in embryonic mouse neural progenitor cells. J Cell Sci. 2002;115:3241–3251. doi: 10.1242/jcs.115.16.3241. [DOI] [PubMed] [Google Scholar]
  20. Ek-Vitorin JF, Calero G, Morley GE, Coombs W, Taffet SM, Delmar M. pH regulation of connexin43: Molecular analysis of the gating particle. Biophys J. 1996;71:1273–1284. doi: 10.1016/S0006-3495(96)79328-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Enkvist MO, McCarthy KD. Astroglial gap junction communication is increased by treatment with either glutamate or high K+ concentration. J Neurochem. 1994;62:489–495. doi: 10.1046/j.1471-4159.1994.62020489.x. [DOI] [PubMed] [Google Scholar]
  22. Franke H, Krugel U, Schmidt R, Grosche J, Reichenbach A, Illes P. P2 receptor-types involved in astrogliosis in vivo. Br J Pharmacol. 2001;134:1180–1189. doi: 10.1038/sj.bjp.0704353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Fumagalli M, Brambilla R, D’Ambrosi N, Volonte C, Matteoli M, Verderio C, Abbracchio MP. Nucleotide-mediated calcium signaling in rat cortical astrocytes: Role of P2X and P2Y receptors. Glia. 2003;43:218-203. doi: 10.1002/glia.10248. [DOI] [PubMed] [Google Scholar]
  24. Giepmans BN. Gap junctions and connexin-interacting proteins. Cardiovasc Res. 2004;62:233–245. doi: 10.1016/j.cardiores.2003.12.009. [DOI] [PubMed] [Google Scholar]
  25. Giepmans BN, Moolenaar WH. The gap junction protein connexin43 interacts with the second PDZ domain of the zona occludens-1 protein. Curr Biol. 1998;8:931–934. doi: 10.1016/s0960-9822(07)00375-2. [DOI] [PubMed] [Google Scholar]
  26. Giepmans BN, Verlaan I, Moolenaar WH. Connexin-43 interactions with ZO-1 and alpha- and beta-tubulin. Cell Commun Adhes. 2001;8:219–223. doi: 10.3109/15419060109080727. [DOI] [PubMed] [Google Scholar]
  27. Grynkiewicz G, Poenie M, Tsien RY. A new generation of Ca2+ indicators with greatly improved fluorescence properties. J Biol Chem. 1985;260:3440–3450. [PubMed] [Google Scholar]
  28. Hinkerohe D, Smikalla D, Haghikia A, Heupel K, Haase CG, Dermietzel R, Faustmann PM. Effects of cytokines on microglial phenotype and astroglial coupling in an inflammatory coculture model. Glia. 2005;52:85–97. doi: 10.1002/glia.20223. [DOI] [PubMed] [Google Scholar]
  29. Horiuchi T, Dietrich HH, Hongo K, Dacey RG., Jr Comparison of P2 receptor subtypes producing dilation in rat intracerebral arterioles. Stroke. 2003;34:1473–1478. doi: 10.1161/01.STR.0000071527.10129.65. [DOI] [PubMed] [Google Scholar]
  30. Iacobas DA, Scemes E, Spray DC. Gene expression alterations in connexin null mice extend beyond the gap junction. Neurochem Int. 2004;45:243–250. doi: 10.1016/j.neuint.2003.12.008. [DOI] [PubMed] [Google Scholar]
  31. Iacobas DA, Suadicani SO, Iacobas S, Chrisman C, Cohen MA, Spray DC, Scemes E. Gap junction and purinergic P2 receptor proteins as a functional unit: Insights from transcriptomics. J Membr Biol. 2007 doi: 10.1007/s00232-007-9039-7. [DOI] [PubMed] [Google Scholar]
  32. Iacobas DA, Urban-Maldonado M, Iacobas S, Scemes E, Spray DC. Array analysis of gene expression in connexin-43 null astrocytes. Physiol Genomics. 2003;15:177–190. doi: 10.1152/physiolgenomics.00062.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Jimenez AI, Castro E, Communi D, Boeynaems JM, Delicado EG, Miras-Portugal MT. Coexpression of several types of metabotropic nucleotide receptors in single cerebellar astrocytes. J Neurochem. 2000;75:2071–2079. doi: 10.1046/j.1471-4159.2000.0752071.x. [DOI] [PubMed] [Google Scholar]
  34. John GR, Scemes E, Suadicani SO, Liu JS, Charles PC, Lee SC, Spray DC, Brosnan CF. IL-1beta differentially regulates calcium wave propagation between primary human fetal astrocytes via pathways involving P2 receptors and gap junction channels. Proc Natl Acad Sci USA. 1999;96:11613–11618. doi: 10.1073/pnas.96.20.11613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Kanemitsu MY, Loo LW, Simon S, Lau AF, Eckhart W. Tyrosine phosphorylation of connexin 43 by v-Src is mediated by SH2 and SH3 domain interactions. J Biol Chem. 1997;272:22824–22831. doi: 10.1074/jbc.272.36.22824. [DOI] [PubMed] [Google Scholar]
  36. Kwak BR, Jongsma HJ. Regulation of cardiac gap junction channel permeability and conductance by several phosphorylating conditions. Mol Cell Biochem. 1996;157:93–99. doi: 10.1007/BF00227885. [DOI] [PubMed] [Google Scholar]
  37. Lampe PD, TenBroek EM, Burt JM, Kurata WE, Johnson RG, Lau AF. Phosphorylation of connexin43 on serine368 by protein kinase C regulates gap junctional communication. J Cell Biol. 2000;149:1503–1512. doi: 10.1083/jcb.149.7.1503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Lee SY, Wolff SC, Nicholas RA, O’Grady SM. P2Y receptors modulate ion channel function through interactions involving the C-terminal domain. Mol Pharmacol. 2003;63:878–885. doi: 10.1124/mol.63.4.878. [DOI] [PubMed] [Google Scholar]
  39. Li W, Hertzberg EL, Spray DC. Regulation of connexin43-protein binding in astrocytes in response to chemical ischemia/hypoxia. J Biol Chem. 2005;280:7941–7948. doi: 10.1074/jbc.M410548200. [DOI] [PubMed] [Google Scholar]
  40. Lin JH, Yang J, Liu S, Takano T, Wang X, Gao Q, Willecke K, Nedergaard M. Connexin mediates gap junction-independent resistance to cell injury. J Neurosci. 2003a;23:430–441. doi: 10.1523/JNEUROSCI.23-02-00430.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Lin D, Zhou J, Zelenka PS, Takemoto DJ. Protein kinase Cgamma regulation of gap junction activity through caveolin-1-containing lipid rafts. Invest Ophthalmol Vis Sci. 2003b;44:5259–5268. doi: 10.1167/iovs.03-0296. [DOI] [PubMed] [Google Scholar]
  42. Lippincott-Schwartz J, Snapp E, Kenworthy A. Studying protein dynamics in living cells. Nat Rev Mol Cell Biol. 2001;2:444–456. doi: 10.1038/35073068. [DOI] [PubMed] [Google Scholar]
  43. Liu J, Liao Z, Camden J, Griffin KD, Garrad RC, Santiago-Perez LI, Gonzalez FA, Seye CI, Weisman GA, Erb L. Src homology 3 binding sites in the P2Y2 nucleotide receptor interact with Src and regulate activities of Src, proline-rich tyrosine kinase 2, and growth factor receptors. J Biol Chem. 2004;279:8212–8218. doi: 10.1074/jbc.M312230200. [DOI] [PubMed] [Google Scholar]
  44. Liu S, Taffet S, Stoner L, Delmar M, Vallano ML, Jalife J. A structural basis for the unequal sensitivity of the major cardiac and liver gap junctions to intracellular acidification: The carboxyl tail length. Biophys J. 1993;64:1422–1433. doi: 10.1016/S0006-3495(93)81508-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Loo LW, Berestecky JM, Kanemitsu MY, Lau AF. pp60src-mediated phosphorylation of connexin 43, a gap junction protein. J Biol Chem. 1995;270:12751–12761. doi: 10.1074/jbc.270.21.12751. [DOI] [PubMed] [Google Scholar]
  46. Matesic DF, Hayashi T, Trosko JE, Germak JA. Upregulation of gap junctional intercellular communication in immortalized gonadotropin-releasing hormone neurons by stimulation of the cyclic AMP pathway. Neuroendocrinology. 1996;64:286–297. doi: 10.1159/000127130. [DOI] [PubMed] [Google Scholar]
  47. Meme W, Calvo CF, Froger N, Ezan P, Amigou E, Koulakoff A, Giaume C. Proinfammatory cytokines released from microglia inhibit gap junctions in astrocytes: Potentiation by beta-amyloid. FASEB J. 2006;20:494–496. doi: 10.1096/fj.05-4297fje. [DOI] [PubMed] [Google Scholar]
  48. Montana V, Ni Y, Sunjara V, Hua X, Parpura V. Vesicular glutamate transporter-dependent glutamate release from astrocytes. J Neurosci. 2004;24:2633–2642. doi: 10.1523/JNEUROSCI.3770-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Morley GE, Taffet SM, Delmar M. Intramolecular interactions mediate pH regulation of connexin43 channels. Biophys J. 1996;70:1294–1302. doi: 10.1016/S0006-3495(96)79686-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Nagy JI, Ionescu AV, Lynn BD, Rash JE. Coupling of astrocyte connexins Cx26, Cx30, Cx43 to oligodendrocyte Cx29, Cx32, Cx47: Implications from normal and connexin32 knockout mice. Glia. 2003;44:205–218. doi: 10.1002/glia.10278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Naus CC, Bond SL, Bechberger JF, Rushlow W. Identification of genes differentially expressed in C6 glioma cells transfected with connexin43. Brain Res Brain Res Rev. 2000;32:259–266. doi: 10.1016/s0165-0173(99)00087-9. [DOI] [PubMed] [Google Scholar]
  52. Neary JT, van Breemen C, Forster E, Norenberg LO, Norenberg MD. ATP stimulates calcium influx in primary astrocyte cultures. Biochem Biophys Res Commun. 1988;157:1410–1416. doi: 10.1016/s0006-291x(88)81032-5. [DOI] [PubMed] [Google Scholar]
  53. Paulson AF, Lampe PD, Meyer RA, TenBroek E, Atkinson MM, Walseth TF, Johnson RG. Cyclic AMP, LDL trigger a rapid enhancement in gap junction assembly through a stimulation of connexin trafficking. J Cell Sci. 2000;113:3037–3049. doi: 10.1242/jcs.113.17.3037. [DOI] [PubMed] [Google Scholar]
  54. Pearce B, Langley D. Purine- and pyrimidine-stimulated phosphoinositide breakdown and intracellular calcium mobilization in astrocytes. Brain Res. 1994;660:329–332. doi: 10.1016/0006-8993(94)91307-2. [DOI] [PubMed] [Google Scholar]
  55. Pina-Benabou MH, Srinivas M, Spray DC, Scemes E. Calmodulin kinase pathway mediates the K+-induced increase in gap junctional communication between mouse spinal cord astrocytes. J Neurosci. 2001;21:6635–6643. doi: 10.1523/JNEUROSCI.21-17-06635.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Rash JE, Yasumura T, Davidson KG, Furman CS, Dudek FE, Nagy JI. Identification of cells expressing Cx43, Cx30, Cx26, Cx32 and Cx36 in gap junctions of rat brain and spinal cord. Cell Commun Adhes. 2001a;8:315–320. doi: 10.3109/15419060109080745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Rash JE, Yasumura T, Dudek FE, Nagy JI. Cell-specific expression of connexins and evidence of restricted gap junctional coupling between glial cells and between neurons. J Neurosci. 2001b;21:1983–2000. doi: 10.1523/JNEUROSCI.21-06-01983.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Rozental R, Morales M, Mehler MF, Urban M, Kremer M, Dermietzel R, Kessler JA, Spray DC. Changes in the properties of gap junctions during neuronal differentiation of hippocampal progenitor cells. J Neurosci. 1998;18:1753–1762. doi: 10.1523/JNEUROSCI.18-05-01753.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Rozental R, Srinivas M, Gokhan S, Urban M, Dermietzel R, Kessler JA, Spray DC, Mehler MF. Temporal expression of neuronal connexins during hippocampal ontogeny. Brain Res Brain Res Rev. 2000;32:57–71. doi: 10.1016/s0165-0173(99)00096-x. [DOI] [PubMed] [Google Scholar]
  60. Saez JC, Connor JA, Spray DC, Bennett MV. Hepatocyte gap junctions are permeable to the second messenger, inositol 1,4,5-trisphosphate, and to calcium ions. Proc Natl Acad Sci USA. 1989;86:2708–2712. doi: 10.1073/pnas.86.8.2708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Saez JC, Martinez AD, Branes MC, Gonzalez HE. Regulation of gap junctions by protein phosphorylation. Braz J Med Biol Res. 1998;31:593–600. doi: 10.1590/s0100-879x1998000500001. [DOI] [PubMed] [Google Scholar]
  62. Scemes E. Components of astrocytic intercellular calcium signaling. Mol Neurobiol. 2000;22:167–179. doi: 10.1385/MN:22:1-3:167. [DOI] [PubMed] [Google Scholar]
  63. Scemes E, Duval N, Meda P. Reduced expression of P2Y+ receptors in connexin43-null mice alters calcium signaling and migration of neural progenitor cells. J Neurosci. 2003;23:11444–11452. doi: 10.1523/JNEUROSCI.23-36-11444.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Scemes E, Giaume C. Astrocytes calcium waves: What they are and what they do. Glia. 2006;54:716–725. doi: 10.1002/glia.20374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Scemes E, Suadicani SO, Spray DC. Intercellular calcium wave communication via gap junction dependent and independent mechanisms. In: Peracchia C, editor. Gap junctions: Molecular basis of cell Communication in health and disease. San Diego, CA: Academic Press; 2000a. pp. 145–173. [Google Scholar]
  66. Scemes E, Suadicani SO, Spray DC. Intercellular communication in spinal cord astrocytes: Fine tuning between gap junctions and P2 nucleotide receptors in calcium wave propagation. J Neurosci. 2000b;20:1435–1445. doi: 10.1523/JNEUROSCI.20-04-01435.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Schubert AL, Schubert W, Spray DC, Lisanti MP. Connexin family members target to lipid raft domains and interact with caveolin-1. Biochemistry. 2002;41:5754–5764. doi: 10.1021/bi0121656. [DOI] [PubMed] [Google Scholar]
  68. Sorgen PL, Duffy HS, Sahoo P, Coombs W, Delmar M, Spay DC. Structural changes in the carboxyl terminus of the gap junction protein connexin43 indicates signaling between binding domains for c-Src and Zonula Occludens-1. J Biol Chem. 2004;279:54695–54701. doi: 10.1074/jbc.M409552200. [DOI] [PubMed] [Google Scholar]
  69. Spray DC, Duffy HS, Scemes E. Gap junctions in glia. Types, roles, and plasticity. Adv Exp Med Biol. 1999;468:339–359. [PubMed] [Google Scholar]
  70. Spray DC, Scemes E. Effects of intracellular pH (and Ca2+) on gap junction channels. In: Kaila K, Ransom BR, editors. pH and brain function. New York: Wiley-Liss; 1998. pp. 477–489. [Google Scholar]
  71. Spray DC, Vink MJ, Scemes E, Suadicani SO, Fishman GI, Dermietzel R. Characteristics of coupling in cardiac myocytes and astrocytes from Cx43(−/−) mice. In: Werner R, editor. Gap junctions. New York: ISO; 1998. pp. 281–285. [Google Scholar]
  72. Striedinger K, Meda P, Scemes E. Exocytosis of ATP from astrocyte progenitors modulates spontaneous Ca2+ oscillations and cell migration. Glia. 2007;55:652–662. doi: 10.1002/glia.20494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Suadicani SO, Flores CE, Urban-Maldonado M, Beelitz M, Scemes E. Gap junction channels coordinate the propagation of intercellular Ca2+ signals generated by P2Y receptor activation. Glia. 2004;48:217–229. doi: 10.1002/glia.20071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Suadicani SO, Pina-Benabou MH, Urban-Maldonado M, Spray DC, Scemes E. Acute downregulation of Cx43 alters P2Y receptor expression levels in mouse spinal cord astrocytes. Glia. 2003;42:160–171. doi: 10.1002/glia.10197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Toyofuku T, Yabuki M, Otsu K, Kuzuya T, Hori M, Tada M. Direct association of the gap junction protein connexin-43 with ZO-1 in cardiac myocytes. J Biol Chem. 1998;273:12725–12731. doi: 10.1074/jbc.273.21.12725. [DOI] [PubMed] [Google Scholar]
  76. Wade MH, Trosko JE, Schindler M. A fluroscence photobleaching assay of gap junction-mediated communication between human cells. Science. 1986;232:525–528. doi: 10.1126/science.3961495. [DOI] [PubMed] [Google Scholar]
  77. Willecke K, Eiberger J, Degen J, Eckardt D, Romualdi A, Guldenagel M, Deutsch U, Sohl G. Structural and functional diversity of connexin genes in the mouse and human genome. Biol Chem. 2002;383:725–737. doi: 10.1515/BC.2002.076. [DOI] [PubMed] [Google Scholar]
  78. Xu X, Li WE, Huang GY, Meyer R, Chen T, Luo Y, Thomas MP, Radice GL, Lo CW. Modulation of mouse neural crest cell motility by N-cadherin and connexin43 gap junctions. J Cell Biol. 2001;154:217–230. doi: 10.1083/jcb.200105047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Zhang Q, Pangrsic T, Kreft M, Krzan M, Li N, Sul JY, Halassa M, Van Bockstaele E, Zorec R, Haydon PG. Fusion-related release of glutamate from astrocytes. J Biol Chem. 2004;279:12724–12733. doi: 10.1074/jbc.M312845200. [DOI] [PubMed] [Google Scholar]
  80. Zhao Y, Rivieccio MA, Lutz S, Scemes E, Brosnan CF. The TLR3 ligand poly I: C downregulates connexin43 expression and function in astrocytes by a mechanism involving the NF-kappaB and PI3 kinase pathways. Glia. 2006;54:775–785. doi: 10.1002/glia.20418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Zhu Y, Kimelberg HK. Developmental expression of metabotropic P2Y1 and P2Y2 receptors in freshly isolated astrocytes from rat hippocampus. J Neurochem. 2001;77:530–541. doi: 10.1046/j.1471-4159.2001.00241.x. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Figure 1S

This article contains supplementary material available via the Internet at http://www.interscience.wiley.com/jpages/0894-1491/suppmat.

Figure 2S
Figure Legends

RESOURCES