Skip to main content
Erschienen in: Journal of Translational Medicine 1/2019

Open Access 01.12.2019 | Research

The important role of connexin 43 in subarachnoid hemorrhage-induced cerebral vasospasm

verfasst von: Le Yang, Jian Yan, Jin-An Zhang, Xin-Hui Zhou, Chao Fang, Er-Ming Zeng, Bin Tang, Jian Duan, Guo-Hui Lu, Tao Hong

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2019

Abstract

Background

Gap junctions are involved in the development of cerebral vasospasm (CVS) after subarachnoid hemorrhage (SAH). However, the specific roles and regulatory functions of related connexin isoforms remain unknown. The aim of this study was to investigate the importance of connexin 43 (Cx43) in CVS and determine whether Cx43 alterations are modulated via the protein kinase C (PKC) signaling transduction pathway.

Methods

Oxyhemoglobin (OxyHb)-induced smooth muscle cells of basilar arterial and second-injection model in rat were used as CVS models in vitro and in vivo. In addition, dye transfer assays were used for gap junction-mediated intercellular communication (GJIC) observation in vitro and delayed cerebral ischemia (DCI) was observed in vivo by perfusion-weighted imaging (PWI) and intravital fluorescence microscopy.

Results

Increase in Cx43 mediated the development of SAH-induced CVS was found in both in vitro and in vivo CVS models. Enhanced GJIC was observed in vitro CVS model, this effect and increased Cx43 were reversed by preincubation with specific PKC inhibitors (chelerythrine or GF 109203X). DCI was observed in vivo on day 7 after SAH. However, DCI was attenuated by pretreatment with Cx43 siRNA or PKC inhibitors, and the increased Cx43 expression in vivo was also reversed by Cx43 siRNA or PKC inhibitors.

Conclusions

These data provide strong evidence that Cx43 plays an important role in CVS and indicate that changes in Cx43 expression may be mediated by the PKC pathway. The current findings suggest that Cx43 and the PKC pathway are novel targets for developing treatments for SAH-induced CVS.
Begleitmaterial
Additional file 1: Fig. S1. Full length blots of Fig. 1A, which include western blotting analysis of Cx43 expression time-course change after OxyHb incubation in vivo. The numbers represent different treatment groups: 1: normal; 2: 24h; 3: 48h; 4: 72h; 5: 96h. Fig. S2. Full length blots of Fig. 1B: Western blotting analysis of the effect of PKC inhibitors on Cx43 protein expression at the time of 48h after OxyHb incubation. The numbers represent different treatment groups: 1: normal; 2: OxyHb-only; 3: OxyHb+CHE; 4: OxyHb+GF; 5: CHE-only; 6: GF-only. Fig. S3. Fluorescence images and corresponding brightfield images in each group. The injected cell is marked with a black arrow. Scale bar = 50 μm. Fig. S4. Blot images of Fig. 3B: Western blotting analysis of Cx43 in BAs derived from the sham and SAH groups. The numbers represent different treatment groups: 1: sham; 2: SAH. Fig. S5. Blot images of Fig. 4A: Western blotting analysis of Cx43 in BAs derived from the non-targeting siRNA (control) or Cx43-targeting siRNA groups after SAH. The numbers represent different treatment groups: 1: control siRNA; 2: Cx43 siRNA. Fig. S6. Blot images of Fig. 4B: Western blotting analysis of Cx43 in BAs derived from the 2 PKC inhibitors groups after SAH. The numbers represent different treatment groups: 1: sham; 2: SAH-only; 3: SAH+CHE; 4: SAH+GF. Fig. S7. Immunolocalization for DAPI and Cx43 in rat subjected to surgery. Tissues were taken 1,3,5 and 14 days after SAH in each group. Scale bar = 5 μm.
Hinweise
Le Yang and Jian Yan contributed equally to this work

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12967-019-02190-1.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
BAs
basilar arteries
BASMCs
basilar arterial smooth muscle cells
CHE
chelerythrine
CVS
cerebral vasospasm
Cx43
connexin 43
DCI
delayed cerebral ischemia
ECs
endothelium cells
EDHF
endothelium-derived hyperpolarizing factor
GF
GF 109203X
GJIC
gap junction intercellular communication
HVJ-E
HVJ envelope
LY
lucifer yellow
MCA
middle cerebral artery
MEGJ
myo-endothelial gap junction
MR PWI
magnet resonance-perfusion weighted imaging
OxyHb
oxyhemoglobin
PKC
protein kinase C
ROIs
regions of interests
rr CBF
relative regional cerebral blood flow
SAH
subarachnoid hemorrhage
SMCs
smooth muscle cells

Introduction

CVS is thought to be a severe complication of SAH. However, the pathogenesis of CVS is not completely understood, and no definitive treatment has been established. Once aneurysm rupture occurred, blood pours into the subarachnoid space even to the brain parenchyma and ventricles. The intracranial pressure rises sharply and might increase enough to affect cerebral perfusion and cause global ischemia. Due to CVS, maximal 7–10 days after onset of SAH, the presence of blood in the subarachnoid spaces triggered and associated with DCI, persistent neurological deficits and long-term neurological disability. DCI is related to the development of CVS, as it is the most important adverse prognostic factor of outcome and a major cause of morbidity and mortality in SAH patients [1]. The pathogenesis of DCI is hypothesized to be multifactorial, including angiographic vasospasm, ischemia, microthrombosis and microcirculation constriction [24]. Due to DCI is among the most important adverse prognostic factors for outcome after SAH [4], it’s of great necessity to explore new targets dealing with the progress of pathology in DCI based on previous SAH animal models [57]. In our previous study [8], we supported the hypothesis that gap junction blockers may relieve the CVS after SAH via cerebral angiography and morphologic study, suggested that gap junctions may play an important role in the pathogenesis of CVS.
Gap junction channels are formed by members of a family of proteins known as connexins [9]. Among them, Cx43 is the most abundant and the major connexin in vascular smooth muscle [10]. Liao Y et al. [11] reported that the conditional knockout of Cx43 in vascular endothelial cells resulted in hypotension, indicating that altered Cx43 expression may be related to irregular vasomotion. In our previous study [8, 12], we reported that Cx43 levels were higher and that vasospasm occurred in the BAs at 7 days after SAH in animals. But, the specific role of Cx43 in DCI after SAH is not well understood.
SAH-induced CVS most often occurs between day 3 and day 7 [1315], indicating that it may be associated with one or more byproducts of blood breakdown in the subarachnoid space, where the exterior surfaces of the cerebral vessels are exposed to blood and byproducts of its breakdown. The breakdown of blood results in the release of OxyHb-derived free radicals, which lead to inhibition of ATP-dependent calcium pumps [16, 17], alterations in vasomotor tone including the release of vasoactive eicosanoids and endothelin from the vessel wall, inhibition of endothelium-dependent relaxation, scavenging of NO and possible influences on the development of CVS [18]. OxyHb-mediated CVS is dependent on the activation of PKC enzymes [19], which are broadly involved in vital cellular functions, including smooth muscle contraction [20].
Several studies have revealed that in vasospastic cerebral arteries, PKC activity is significantly enhanced in the membrane fraction [21], and the time courses of the progression of CVS and PKC activation are well correlated, as revealed by an enzyme immunoassay study [22]. However, the specific mechanism of PKC activation leading to CVS is unclear. Joshi CN et al. [23] demonstrated an important role of Cx43 in the proliferation of vascular SMCs: increased Cx43 expression was significantly reversed in the presence of PKA, PKG and PKC inhibitor in thoracic aorta SMCs. In addition, S Nishizawa et al. proposed that PKC isoforms potentially play a role in the initiation and maintenance of delayed CVS [24] implicating the PKC pathway in Cx43 expression and Cx43-mediated GJIC. But, to date, there is no information available regarding the possible involvement of PKC in Cx43 alterations and gap junction remodeling on cerebrovascular SMCs under spastic conditions. The purpose of the present study was to determine the actual role of Cx43 on DCI after SAH and its involvement in the PKC pathway to lay the groundwork for developing therapeutic strategies for SAH.
In the current study, the time-course changes in Cx43 both in vitro and in vivo were explored. The exact location of Cx43 expression in BA walls was detected using a laser-scanning confocal microscope. By specifically knocking down Cx43 in BAs using siRNA interference, we correlated the important role of Cx43 with DCI in a common SAH model. In addition, by designing an experimental therapeutic study using specific PKC inhibitors, we provide evidence that Cx43-mediated GJIC enhancement and DCI may be modulated via the PKC pathway, which could be used as reference data for developing and analyzing neuroprotective strategies in further studies.

Materials and methods

Rat cell culture and SAH model established in vitro

Rat BASMCs were isolated by enzymatic digestion as previously described [25]. As a major component of blood, OxyHb has been proven to be the principal cause of CVS and delayed neurological deficits following aneurismal SAH. It has also been used to induce the SAH model in vitro as described previously [26]. To mimic SAH in vitro, SMCs were exposed to OxyHb at 10−6 M with complete culture medium in all dishes. SMCs passaged for 3 generations were cultured to confluence. The total medium was then discarded. After rinsing twice with sterile PBS, complete culture medium with OxyHb was added. The dishes were then separated into four groups incubated with OxyHb for different times: 24 h in Group I, 48 h in Group II, 72 h in Group III, and 96 h in Group IV. To study the effects of PKC inhibition, Group II was further divided into three subgroups corresponding to a OxyHb-only group and OxyHb groups pretreated with two specific PKC inhibitors: (i) chelerythrine (CHE) [27] and (ii) GF 109203X (GF) [28]. The total protein of all the cells were collected and stored at − 80 °C until analysis of Cx43 via Western blot. All animal procedures were carried out in accordance with the Guide for the Care and Use of Laboratory Animals published by the US National Institutes of Health (NIH publication No. 86-23, revised 1985); animal use and welfare followed a protocol reviewed and approved by the First Affiliated Hospital of Nanchang University.

Dye transfer assays

The intercellular dye transfer assay of Lucifer yellow (LY) (5% w/v in 150 mM LiCl) was evaluated by microinjecting the dye into a single BASMC and evaluating the diffusion of the dye into neighboring cells in a cell monolayer, as previously described [29]. Single cells were microinjected using a Picoliter Injector (model PLI-90, Harvard Apparatus, South Natick, MA, USA) containing the dye until the impaled cell was brightly fluorescent. 5 min. after injection, the cells were photographed by epifluorescence and the extent of intercellular transfer of LY was determined by counting the number of adjacent cells containing the dye tracer [30]. Experiments were repeated in triplicate per experiment.

Induction of experimental SAH and experimental therapeutic study

Adult male Sprague–Dawley rats were provided by the Department of Laboratory Animal Science, Medical College of Nanchang University, Nanchang, China. The double-hemorrhage model of SAH used here was described previously [31]. Briefly, on both day 1 and 2, the rats were anesthetized using 3% isoflurane in an oxygen and nitrous oxide mixture (1:2). After the loss of the righting reflex, the rat was transferred to a heating blanket (Harvard Apparatus Ltd., UK) coupled to a rectal probe for the maintenance of body temperature (37.5 ± 0.5 °C). After a small incision in the atlanto-occipital membrane, 0.1 ml cerebrospinal fluid was withdrawn, and 0.2 ml non-heparinized blood was injected through the needle inserted into the cisterna magna. The head of the rat was placed in a nose-downward position for 20 min to distribute the blood into subarachnoid spaces after second injection. In a control “sham” group, the atlanto-occipital membrane was exposed but equivalent normal saline was injected to determine if the surgery significantly altered the outcome of the current study. The animals were euthanized by overdose with carbon dioxide (CO2) gas 1, 3, 5, 7 and 14 days after SAH induction, and the BAs were removed for Western blotting or immunohistochemistry.
Knockdown of Cx43 was performed by using an HVJ envelope (HVJ-E) transfection kit (GenomONE™, Cosmo Bio Co., Ltd., Tokyo, Japan), which is widely used for in vivo siRNA transfer [32, 33]. Cx43 siRNA (siGENOME SMARTpool, mouse GJA1, Thermo Scientific) [34, 35] was incorporated into the HVJ-E according to the manufacturer’s instructions. Two days prior to SAH induction, siRNA (5 pg in 10 μl sterile saline) was slowly injected into the cisterna magna as pretreatment, similar to the induction of SAH described above. siGENOME Non-Targeting siRNA Control Pool #2 (Thermo Scientific) was used as a negative control. In addition, the pharmacological effects of the two PKC inhibitors were examined in separate groups. Two days prior to SAH induction, 3 ml sterile PBS containing 5 μM CHE or GF was injected into the cisterna magna, and on day 3 and day 5 after SAH induction, we injected the same amount of each drug into the cisterna magna. Seven days after SAH induction, animals were reanesthesized to perform MR PWI or intravital fluorescence microscopy. After completion of these examinations, the BAs were removed for Western blotting or immunohistochemistry.

MRI protocol and intravital fluorescence microscopy of rats

After SAH induction, the rats were randomly divided in two groups, which were respectively examined by functional MRI and intravital fluorescence microscopy on day 7, the day of the maximum CVS and DCI, as previous reported [36] with maximum vasospasm detected after day 7 using transcranial Doppler ultrasound. Each group was further divided into six subgroups as follows: (I) a sham-operated group, (II) an SAH-only group, (III) an SAH + Cx43siRNA group, (IV) an SAH + control siRNA group, (V) an SAH + CHE group and (VI) an SAH + GF group. For postanesthetic animals, MR PWI experiments were carried out using a 7.0T/30-cm imaging system (Bruker Biospec, Germany) with a gradient inset (121 mm internal diameter, 400 mT/m). For bolus tracking PWI of axial slices, a magnetic resonance gradient echo sequence was used near the position of hippocampus. After standardized intravenous injection of 0.2 ml Gd-DTPA (0.1 mmol/kg) at a flow rate of 0.15 ml/s, 22 sequential images were obtained in approximately 60 s. PWI post-processing was implemented with Signal Processing In Nmri software (SPIN, MRI Institute for Biomedical Research, Detroit, MI, USA). Relative regional (rr) blood flow (BF) was calculated semiquantitatively in different ROIs. In each animal, ROIs were selected in the hippocampus and the masseter muscle. The ratio of rr CBF to rr masseter muscle BF was defined to determine the reduction of CBF due to CVS [37]. For each group, a mean value of the ratio was calculated. In another group, the animals underwent an intravital fluorescence microscopy experiment.
For intravital fluorescence microscopy, each subgroup was assessed with intravital microscopy according to a previous study [3840]. The methods of Friedrich B, et al. [38] with slight modification were used for microvessel assessment. Briefly, vessel diameters were quantified in the images captured by video and analyzed using IC Viewer software (Mauna Kea Technologies, Paris, France). The percentage of spastic vessels among all vessels in each group were calculated. Individual vessel constrictions were calculated by dividing the diameter of the most constricted vessel segments (Fig. 5a, white arrow) by the diameter of the nearest non-constricted vessel segment among each group. To confirm that non-constricted vessel segments indeed represent the baseline diameter of the vessel, microvessels were categorized by their degree of branching from the MCA (A1 to A6). The mean diameter of each vessel category was calculated and compared between sham-operated rats and SAH rats. No difference was detected between the groups, indicating that the non-constricted vessel segments in the rats subjected to SAH retained physiological baseline characteristics.

Western blot analysis

Western blot analysis was used to analyze changes in the expression of Cx43 protein. Briefly, proteins from each group of BAs or SMCs were subjected to SDS-PAGE and then transferred to nitrocellulose membranes, which were blocked for 2 h in Tris-buffered saline Tween (TBST) containing 5% milk powder at room temperature. The anti-Cx43 antibody (MAB3067; Sigma, diluted 1:1000) and anti-GAPDH antibody (AB2302; Sigma, diluted 1:1000) were used to the membrane in saturation buffer. After extensively washing 5 times in TBST for 10 min each, the membranes were probed for 2 h with horseradish peroxide-conjugated goat anti-rabbit secondary antibody (diluted 1:2000 in saturation buffer). The durations of exposure to hyperfilm and incubation in ECL detection reagents were maintained consistent for all conditions. The intensity of the bands after Western blotting was determined by laser scanning of the films, followed by quantitative densitometric analysis using Kodak Digital Science 1D 2.0 Image software, and the protein expression levels of Cx43 were normalized to GAPDH.

Statistical analysis

Data are presented as the mean ± SEM. Statistical significance of differences were assessed by one-way analysis of variance (ANOVA) followed by Student’s t-tests using IBM SPSS Statistics, Version 21. The results were considered significant for p < 0.05.

Results

In vitro detection of Cx43 protein levels in the experimental vasospasm model among cultured BASMCs and the involvement of the PKC pathway

As shown in Fig. 1a of different groups, we observed that OxyHb (10−6 M) incubation in different time points, the Cx43 protein expression gradually increased with a maximal at 48 h, and then returned to control by 96 h. Exposure of the cells to CHE or GF alone had no effect on the level of Cx43 protein expression compared to normal cells, but both CHE and GF significantly prevented the maximal expression(48 h) in Cx43, suggesting a role of PKC in OxyHb-induced Cx43 expression (Fig. 1b).

The effect of PKC inhibition on OxyHb-induced enhancement of gap junction intercellular communication in BASMCs

Normally, an average of 9.88 ± 2.59 cells coupled to each cell was detected. Incubation with OxyHb for 48 h significantly increased the numbers of coupled cells to 18.5 ± 3.12 cells per microinjection (Fig. 2a, b, dye coupling index). As expected, both PKC inhibitors significantly reduced Lucifer yellow transfer by more than 20% (14.13 ± 2.70 and 13.88 ± 2.42, respectively, P < 0.05) when OxyHb was added with the pretreatment of CHE or GF. However, CHE or GF alone did not affect the migration of LY between cells compared with controls, which corresponded with our observation of Cx43 levels.

In vivo experiment of the time-course changes in Cx43 protein levels among BAs

In the SAH group, the blood was observed diffusely distributed in the subarachnoid space and around Willis’ circle on day 7 (Fig. 3a). In the sham group, no significant change in Cx43 was found throughout the time course after surgery. However, similar to the in vitro findings, Cx43 alterations in the BAs were also time dependent in the SAH group compared to the corresponding time periods in the sham group in vivo. Cx43 was significantly increased from the first day and continuously elevated to a peak value at 7 days following SAH; 14 days after SAH induction, the expression levels of Cx43 were similar in the sham and control SAH groups (Fig. 3b) which further verified our hypothesis. In addition, immunohistochemistry was performed to identify the cell type that expresses Cx43. In the sham-operated group (Fig. 3c, upper panel), Cx43 was colocalized with DAPI, and Cx43-stained punctate was abundant around the smooth muscle cells of all the BAs examined, suggesting pericellular localization. In the media, discrete spots were distributed circumferentially around the vessel wall, mimicking the orientation of the smooth muscle cells, which is consistent with previous findings [41]. However, beneath the internal elastic lamina, it was difficult to distinguish the staining around the ECs (Fig. 3c, white arrow). Staining was rarely observed around the ECs in the SAH group, similar with the sham-operated group. This finding suggests that in BAs, Cx43 is mainly expressed in the SMC layer rather than in the ECs. Consistent with the Western blotting findings, compared with the Cx43 immunofluorescence intensity of the sham-operated group, that of the SAH group increased gradually from 1 to 3 and 5 days after SAH and then peaked on day 7 (Fig. 3c, lower panel). Fourteen days after SAH, Cx43 immunofluorescence intensity had returned to normal levels (Additional file 1: Fig. S7). Hence, our hypothesis was further verified.

Effects of Cx43 siRNA and PKC inhibitors on the experimental therapeutic group of rats

Cx43 expression gradually increased to day 7 and then decreased to normal levels by day 14 in the control siRNA group with SAH induction. In striking contrast, no significant elevation was detected along the same time course in the Cx43 siRNA group. In comparison with the levels at the corresponding time periods in the control siRNA group, the efficacy of Cx43 attenuation was observed beginning at day 3 after cisterna magna injection of Cx43-targeting siRNA. On day 3, 5 and day 7 in the Cx43 siRNA group, the expression of Cx43 tended to be significantly lower than that of the non-targeting siRNA group (Fig. 4a). In another experimental therapeutic group, the peak increased expression of Cx43 in the BAs at day 7 post-SAH was reversed by using either PKC inhibitor, CHE or GF (Fig. 4b).
Furthermore, immunohistochemistry revealed that Cx43 expression in the SMC layer was reduced 7 days after the injection of Cx43 siRNA compared with that of the non-targeting siRNA (Fig. 4c, upper panels). Immunohistochemistry also showed that Cx43 expression in the SMC layer was reduced in the two SAH + PKC inhibitor groups (Fig. 4c, lower panels) compared with that in the SAH-only group (Fig. 3c); this finding verified that PKC inhibitors reverse the increase in Cx43 expression.

Knockdown of Cx43 via siRNA and PKC inhibitors leads to remission of vasospasm and reversal of CBF after SAH in rats

No microvascular constrictions were observed in sham-operated rats, and numerous constricted microvessels were observed at 7 days after SAH. As expected, the vasospastic pattern is similar to the pearl string-like constrictions observed in SAH patients. Quantification of these observations (Fig. 5b) revealed that, at 7 days after SAH, a significantly increased proportion (P < 0.01 vs. sham group, n = 8) of vessel segments derived from the MCA showed one or more constrictions in the SAH-only (61.33 ± 6.87%) and SAH + control siRNA (60.83 ± 5.12%) groups. Nevertheless, though an increased proportion (P < 0.01 vs. sham group, n = 8) of constricted vessel segments was also observed in the SAH + Cx43 siRNA group (51.28 ± 8.37%), knockdown of Cx43 via siRNA decreased the proportion of constricted vessel segments compared with the SAH-only group (P < 0.05). In parallel with the Western blot findings, the two SAH + PKC inhibitor groups also showed decreased proportions to different extents, suggesting the important role of Cx43 in microvascular spasms after SAH and the involvement of the PKC pathway. In addition, the severity of microvessel constriction was assessed by changes in microvessel diameters (Fig. 5c). Seven days after SAH, the diameters of affected vessel segments were significantly decreased in the SAH-only and SAH + control siRNA groups compared with the sham group (P < 0.01). In the SAH + Cx43 siRNA group and the two SAH + PKC inhibitor groups, the severity of constriction led to remission compared with the SAH-only group (P < 0.01). These data indicated increases in both the proportion and severity of microvasospasms at 7 days after SAH, which could be alleviated by knockdown of Cx43 via RNA interference and PKC inhibition.
rrCBF was measured via MR PWI, which is a general semiquantitative method, and the contralateral hemisphere was used as a reference [4244]. However, in SAH-induced CVS, the reduction of CBF is not considered to be unilateral [45, 46]. So in the present study, blood flow of the masseter muscle was used as a reference [37]. According to previous studies, the CBF/masseter muscle BF ratio in male Sprague–Dawley rats was reported within a range of 6.1–7.7. Thus, the rr CBF/rr masseter muscle BF ratio may serve as a good quantitative indicator for analyzing reduced CBF. Here, rrCBF was 7.25 ± 1.37-fold higher compared to the perfusion of the rr masseter muscle in sham-operated rats, which is consistent with previous studies [47, 48]. However, the ratio was reduced to 3.16 ± 1.23 on day 7 after SAH. In our previous study [8] we demonstrated that Cx43 increased, and vasospasms occurred in SAH animals. Together with the observation above that Cx43 was significantly increased on day 7, our data demonstrate a significant correlation between the defined ratio (CBF/muscular BF) and the increased Cx43 expression 7 days after SAH with DCI. Additionally, on day 7 post-SAH, both the SAH + Cx43 siRNA and SAH + PKC inhibitor (CHE or GF) groups exhibited a significantly smaller reduction in CBF, while there was no significant change in the SAH + control siRNA group (Fig. 6).

Discussion

Our present study showed that Cx43 protein was increased in the in vitro CVS model with OxyHb incubation. Moreover, our dye transfer assay also revealed enhanced GJIC in BASMCs in culture incubated with OxyHb. Our previous study demonstrated that Cx43 expression may up-regulate contractions in rabbit BAs via a GJIC-dependent mechanism evaluated by a scrape-loading method [49], we further confirmed here. The enhancement in GJIC may lead to exaggerated contractile responses among more coupled SMCs. Thus, spasmogenic signals such as red blood cell breakdown and the metabolic products spreading into the subarachnoid space could propagate more rapidly and forcefully from the ruptured vessel wall to a more extensive area, causing a prolonged contractile response. In addition, in our previous study, we showed that heptanol, a gap junction inhibitor, significantly inhibited the sustained contraction of rabbit BA rings induced by OxyHb in a dose-dependent manner [12]. The implication of gap junctions, especially Cx43, in OxyHb-induced CVS was further demonstrated here.
Both specific inhibitors of PKC we selected (CHE/GF) have been used to study the involvement of PKC in signal transduction pathways [27, 28]. By treatment with the two PKC inhibitors, the increased Cx43 protein and the enhanced GJIC were both reversed, implying that OxyHb-induced Cx43 alterations and enhanced GJIC may be modulated via the PKC pathway. However, previous studies have reported that PKC is composed of a family of serine–threonine kinases consisting of three major classes with at least 11 different isoforms [50]. During the pathological process of canine basilar artery cells in previous CVS research [24], PKCδ translocates from the cytosol to the membrane in early stage, and PKCα is subsequently translocated, suggesting that PKCδ plays a role during initiation and PKCα in the maintenance of CVS. However, as the two PKC inhibitors used here are both non-selective, the specific isoforms of PKC and their functional roles leading to Cx43 alterations are presently unknown and their identity remains to be elucidated. Furthermore, Richards et al. [51] proposed that PKC spatially and temporally controls GJIC via phosphorylation of Cx43 at serine 368, meanwhile as PKC activity was significantly enhanced in the membrane fraction of vasospastic cerebral arteries, the enhanced GJIC revealed here may be associated with the phosphorylation level of Cx43 at serine 368. Interestingly, given that the total expression of Cx43 protein also increased significantly in our observations, it seems that the modulation of GJIC may result from the ratio of pS368-Cx43 to total Cx43, which remains to be explored.
In our previous study [8], we found that vasospasm was most intense and coincidentally that the expression of Cx43 peaked on day 7. Together with our observation of the time phase change of Cx43 on SMCs, our findings further determined the involvement of increased Cx43 in SAH-induced CVS both in vivo and in vitro. On the other hand, DCI is thought to be caused by the combined effects of cortical spreading ischemia, arteriolar constriction and thrombosis. Here, DCI was investigated by performing intravital fluorescence microscopy and MRI measurements by PWI in rats. We directly visualized the post-SAH microcirculation in vivo by intravital fluorescence microscopy. This technique is advantageous over ex vivo studies for investigating arterioles in the living brain, allowing us to observe vasospasms and dynamic interactions in the entire vascular tree of MCA. Our main findings of the DCI investigation were that CBF significantly decreased and over 60% of arterioles derived from the MCA showed pearl string-like constrictions on day 7 after SAH. As reducing vessel diameter by 30% could reduce flow by ~ 80%, leading to DCI, our results may explain why DCI is observed after SAH in both experimental animals and patients [5153]. It suggesting that changes in Cx43 might be implicated in the pathophysiological events of DCI since both DCI and increment of Cx43 coincidentally occurred on day 7.
After knockdown of Cx43 via RNA interference, MCA deficiency and reduced CBF were both eased while control siRNA did not significantly relieve the deficits. In a previous study [54], corrosion casts of arterioles showed that they exhibited tapered narrowing with folding after SAH. Additionally, the width of the arterioles was significantly lower at 3 and 7 days after SAH. Morphometric examination by light microscopy showed that the internal diameter of the arterioles was significantly smaller and that this change was associated with a significant increase in wall thickness at any depth from the brain at 7 days after SAH. This condition had improved by 14 days after SAH, suggesting that arteriole constriction occurs after SAH and may contribute to DCI. A previous study [55] used orthogonal polarization spectral (OPS) imaging and found that in SAH patients, capillary density was significantly lower and the small arteries and arterioles at the cortical surface exhibited vasospasm that was not detected by angiography or transcranial Doppler sonography. These changes indicated that CVS may be associated with a significant decrease in capillary perfusion. Although Cx43 expression was restored by pretreatment with PKC inhibitors or Cx43 siRNA, resulting in the significant alleviation of the deleterious effects of SAH, the microvascular spasms were not completely reversed (Fig. 5). In our Cx43 siRNA interference experiment (Fig. 4a), the efficacy reached an ideal level prior to days 3–5. Because the development of constriction in microarterioles is a gradual process and because a previous study in rats [56] showed that subarachnoid arterioles constrict by ~ 40% in the early phase, i.e., 5 min to 2 h after SAH in vivo, we suspect that the efficacy of Cx43 siRNA interference and the occurrence of micro-constriction are not synchronously correlated. Thus, in accordance with our proposed hypothesis, we showed that the severity of CVS and, subsequently, DCI induced by SAH were strongly related to vascular Cx43 alterations. Moreover, in addition to SMC, astrocytes are also important regulators of vasomotor reaction. The high abundance of gap junctions in astrocytes allows for the direct intercellular diffusion of ions, nutrients, and signaling molecules between these cells. Cx43 is the most abundant connexin expressed in astrocytes and thus constitutes the major connexin contributing to gap junction communication in astrocytes. Hence, SAH-induced alterations in the expression and functions of Cx43 should be systematically explored in other types of cells in future studies.
Endothelium-mediated vasodilatation is evoked by increased calcium concentrations in the endothelium, which triggers hyperpolarization by activating endothelial calcium-dependent potassium channels [57], leading to the recognition of endothelium-dependent hyperpolarizing factor (known as EDHF). Thus, the conduction of EDHF via MEGJs to the underlying smooth muscle cell layer results in a decrease in intracellular calcium and subsequent vasodilatation. Because vasodilation signals upon focal stimulation of arterioles have been shown to be conducted along the vascular wall [58], Hoepfl B et al. [59] suggested that EDHF, but not NO or prostaglandins, serves as a critical mediator to initiate conducted vasodilation (CVD) upon acetylcholine administration in hamster arterioles. In the present study, Cx43 increased significantly after SAH, and it is believed that progressive up-regulation of Cx43 in SMCs may result in the development of intimal hyperplasia [60], and increasing intimal hyperplasia mechanically dissociates endothelial–medial interactions at the MEGJ interface. Decreasing myoendothelial interactions progressively attenuates the hyperpolarization that normally passes from the ECs to medial SMCs, eventually leading to irregular vasomotion. Moreover, studies using connexin-mimetic peptides to selectively inhibit GJIC in rabbit iliac arteries suggested that Cx43 is required for propagation of EDHF within the smooth muscle layer [61], implying the involvement of Cx43 in disruption of CVD. Although our data demonstrated that Cx43 is closely related to the CVS in pathological conditions of SAH, these findings do not explain the specific mechanism of Cx43 underlying the impaired vasodilation. Although SMCs have been reported to primarily express Cx43 [41] and ECs to primarily express Cx40 [62, 63], the connexin content of MEGJs remains unknown and has not been adequately explored. In contrast with Cx43 knockout mice, constitutive deletion of Cx40 results in hypertension in both anesthetized and awake mice [64, 65]. Cx40 is also critical for transferring vasodilatory signals from ECs to the vascular media [66]. Moreover, it has been proposed that nearly a twofold difference in the Cx43:Cx40 expression ratio gives rise to more than a 25-fold difference in dye coupling via the GJIC in vitro [67]. Together with our observation that Cx43/Cx40 proteins form heteromeric gap junctions that are increased in SAH, we hypothesize that the Cx43:Cx40 ratio maintains a certain pattern for the maintenance of various physiological functions under normal conditions. Once SAH occurred, the connexin patterns varied, and both increased Cx43 and decreased Cx40 within the cerebral vessel wall may be responsible for the development of CVS. The present findings suggest a novel emphasis on this research direction for future studies.

Conclusion

In summary, the important role of Cx43 in SAH-induced CVS was affirmed in present study. Gap junction remodeling occurred after SAH and may be implicated in irregular vasomotion of CVS. Moreover, increased Cx43 after SAH may be modulated via the PKC pathway.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12967-019-02190-1.

Acknowledgements

Not applicable.
All animal procedures were carried out in accordance with the Guide for the Care and Use of Laboratory Animals published by the US National Institutes of Health (NIH publication no. 86-23, revised 1985); animal use and welfare followed a protocol reviewed and approved by the First Affiliated Hospital of Nanchang University.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Supplementary information

Additional file 1: Fig. S1. Full length blots of Fig. 1A, which include western blotting analysis of Cx43 expression time-course change after OxyHb incubation in vivo. The numbers represent different treatment groups: 1: normal; 2: 24h; 3: 48h; 4: 72h; 5: 96h. Fig. S2. Full length blots of Fig. 1B: Western blotting analysis of the effect of PKC inhibitors on Cx43 protein expression at the time of 48h after OxyHb incubation. The numbers represent different treatment groups: 1: normal; 2: OxyHb-only; 3: OxyHb+CHE; 4: OxyHb+GF; 5: CHE-only; 6: GF-only. Fig. S3. Fluorescence images and corresponding brightfield images in each group. The injected cell is marked with a black arrow. Scale bar = 50 μm. Fig. S4. Blot images of Fig. 3B: Western blotting analysis of Cx43 in BAs derived from the sham and SAH groups. The numbers represent different treatment groups: 1: sham; 2: SAH. Fig. S5. Blot images of Fig. 4A: Western blotting analysis of Cx43 in BAs derived from the non-targeting siRNA (control) or Cx43-targeting siRNA groups after SAH. The numbers represent different treatment groups: 1: control siRNA; 2: Cx43 siRNA. Fig. S6. Blot images of Fig. 4B: Western blotting analysis of Cx43 in BAs derived from the 2 PKC inhibitors groups after SAH. The numbers represent different treatment groups: 1: sham; 2: SAH-only; 3: SAH+CHE; 4: SAH+GF. Fig. S7. Immunolocalization for DAPI and Cx43 in rat subjected to surgery. Tissues were taken 1,3,5 and 14 days after SAH in each group. Scale bar = 5 μm.
Literatur
2.
Zurück zum Zitat Macdonald RL. Delayed neurological deterioration after subarachnoid haemorrhage. Nat Rev Neurol. 2014;10(1):44–58.PubMedCrossRef Macdonald RL. Delayed neurological deterioration after subarachnoid haemorrhage. Nat Rev Neurol. 2014;10(1):44–58.PubMedCrossRef
3.
Zurück zum Zitat Rosengart AJ, Huo JD, Tolentino J, Novakovic RL, Frank JI, Goldenberg FD, et al. Outcome in patients with subarachnoid hemorrhage treated with antiepileptic drugs. J Neurosurg. 2007;107(2):253–60.PubMedCrossRef Rosengart AJ, Huo JD, Tolentino J, Novakovic RL, Frank JI, Goldenberg FD, et al. Outcome in patients with subarachnoid hemorrhage treated with antiepileptic drugs. J Neurosurg. 2007;107(2):253–60.PubMedCrossRef
4.
Zurück zum Zitat Rosengart AJ, Schultheiss KE, Tolentino J, Macdonald RL. Prognostic factors for outcome in patients with aneurysmal subarachnoid hemorrhage. Stroke. 2007;38(8):2315–21.PubMedCrossRef Rosengart AJ, Schultheiss KE, Tolentino J, Macdonald RL. Prognostic factors for outcome in patients with aneurysmal subarachnoid hemorrhage. Stroke. 2007;38(8):2315–21.PubMedCrossRef
5.
Zurück zum Zitat Lin CL, Calisaneller T, Ukita N, Dumont AS, Kassell NF, Lee KS. A murine model of subarachnoid hemorrhage-induced cerebral vasospasm. J Neurosci Methods. 2003;123(1):89–97.PubMedCrossRef Lin CL, Calisaneller T, Ukita N, Dumont AS, Kassell NF, Lee KS. A murine model of subarachnoid hemorrhage-induced cerebral vasospasm. J Neurosci Methods. 2003;123(1):89–97.PubMedCrossRef
6.
Zurück zum Zitat El Amki M, Dubois M, Lefevre-Scelles A, Magne N, Roussel M, Clavier T, et al. Long-lasting cerebral vasospasm, microthrombosis, apoptosis and paravascular alterations associated with neurological deficits in a mouse model of subarachnoid hemorrhage. Mol Neurobiol. 2018;55(4):2763–79.PubMedCrossRef El Amki M, Dubois M, Lefevre-Scelles A, Magne N, Roussel M, Clavier T, et al. Long-lasting cerebral vasospasm, microthrombosis, apoptosis and paravascular alterations associated with neurological deficits in a mouse model of subarachnoid hemorrhage. Mol Neurobiol. 2018;55(4):2763–79.PubMedCrossRef
7.
Zurück zum Zitat Ciurea AV, Palade C, Voinescu D, Nica DA. Subarachnoid hemorrhage and cerebral vasospasm—literature review. J Med Life. 2013;6(2):120–5.PubMedPubMedCentral Ciurea AV, Palade C, Voinescu D, Nica DA. Subarachnoid hemorrhage and cerebral vasospasm—literature review. J Med Life. 2013;6(2):120–5.PubMedPubMedCentral
8.
Zurück zum Zitat Hong T, Wang H, Wang Y, Wang H. Effects of gap junctional blockers on cerebral vasospasm after subarachnoid hemorrhage in rabbits. Neurol Res. 2009;31(3):238–44.PubMedCrossRef Hong T, Wang H, Wang Y, Wang H. Effects of gap junctional blockers on cerebral vasospasm after subarachnoid hemorrhage in rabbits. Neurol Res. 2009;31(3):238–44.PubMedCrossRef
9.
Zurück zum Zitat Beyer EC, Paul DL, Goodenough DA. Connexin family of gap junction proteins. J Membr Biol. 1990;116(3):187–94.PubMedCrossRef Beyer EC, Paul DL, Goodenough DA. Connexin family of gap junction proteins. J Membr Biol. 1990;116(3):187–94.PubMedCrossRef
10.
Zurück zum Zitat Brink PR, Ricotta J, Christ GJ. Biophysical characteristics of gap junctions in vascular wall cells: implications for vascular biology and disease. Braz J Med Biol Res. 2000;33(4):415–22.PubMedCrossRef Brink PR, Ricotta J, Christ GJ. Biophysical characteristics of gap junctions in vascular wall cells: implications for vascular biology and disease. Braz J Med Biol Res. 2000;33(4):415–22.PubMedCrossRef
11.
Zurück zum Zitat Liao Y, Day KH, Damon DN, Duling BR. Endothelial cell-specific knockout of connexin 43 causes hypotension and bradycardia in mice. Proc Natl Acad Sci USA. 2001;98(17):9989–94.PubMedCrossRefPubMedCentral Liao Y, Day KH, Damon DN, Duling BR. Endothelial cell-specific knockout of connexin 43 causes hypotension and bradycardia in mice. Proc Natl Acad Sci USA. 2001;98(17):9989–94.PubMedCrossRefPubMedCentral
12.
Zurück zum Zitat Hong T, Wang Y, Wang HT, Wang H. Inhibitory effect of gap junction blockers on cerebral vasospasm. J Neurosurg. 2008;108(3):551–7.PubMedCrossRef Hong T, Wang Y, Wang HT, Wang H. Inhibitory effect of gap junction blockers on cerebral vasospasm. J Neurosurg. 2008;108(3):551–7.PubMedCrossRef
13.
Zurück zum Zitat Dorsch NW. Therapeutic approaches to vasospasm in subarachnoid hemorrhage. Curr Opin Crit Care. 2002;8(2):128–33.PubMedCrossRef Dorsch NW. Therapeutic approaches to vasospasm in subarachnoid hemorrhage. Curr Opin Crit Care. 2002;8(2):128–33.PubMedCrossRef
14.
Zurück zum Zitat Gerard E, Frontera JA, Wright CB. Vasospasm and cerebral infarction following isolated intraventricular hemorrhage. Neurocrit Care. 2007;7(3):257–9.PubMedCrossRef Gerard E, Frontera JA, Wright CB. Vasospasm and cerebral infarction following isolated intraventricular hemorrhage. Neurocrit Care. 2007;7(3):257–9.PubMedCrossRef
15.
Zurück zum Zitat Haley EC, Kassell NF, Torner JC. The international cooperative study on the timing of aneurysm surgery. North Am Exp Stroke. 1992;23(2):205–14. Haley EC, Kassell NF, Torner JC. The international cooperative study on the timing of aneurysm surgery. North Am Exp Stroke. 1992;23(2):205–14.
16.
Zurück zum Zitat Arai T, Takeyama N, Tanaka T. Glutathione monoethyl ester and inhibition of the oxyhemoglobin-induced increase in cytosolic calcium in cultured smooth-muscle cells. J Neurosurg. 1999;90(3):527–32.PubMedCrossRef Arai T, Takeyama N, Tanaka T. Glutathione monoethyl ester and inhibition of the oxyhemoglobin-induced increase in cytosolic calcium in cultured smooth-muscle cells. J Neurosurg. 1999;90(3):527–32.PubMedCrossRef
17.
Zurück zum Zitat Macdonald RL, Weir BK, Marton LS, Zhang ZD, Sajdak M, Johns LM, et al. Role of adenosine 5′-triphosphate in vasospasm after subarachnoid hemorrhage: human investigations. Neurosurgery. 2001;48(4):854–63 Discussion 862–3.PubMed Macdonald RL, Weir BK, Marton LS, Zhang ZD, Sajdak M, Johns LM, et al. Role of adenosine 5′-triphosphate in vasospasm after subarachnoid hemorrhage: human investigations. Neurosurgery. 2001;48(4):854–63 Discussion 862–3.PubMed
18.
Zurück zum Zitat Macdonald RL, Weir BK. A review of hemoglobin and the pathogenesis of cerebral vasospasm. Stroke. 1991;22(8):971–82.PubMedCrossRef Macdonald RL, Weir BK. A review of hemoglobin and the pathogenesis of cerebral vasospasm. Stroke. 1991;22(8):971–82.PubMedCrossRef
19.
Zurück zum Zitat Wickman G, Lan C, Vollrath B. Functional roles of the Rho/Rho kinase pathway and protein kinase C in the regulation of cerebrovascular constriction mediated by hemoglobin. Circ Res. 2003;92:809–16.PubMedCrossRef Wickman G, Lan C, Vollrath B. Functional roles of the Rho/Rho kinase pathway and protein kinase C in the regulation of cerebrovascular constriction mediated by hemoglobin. Circ Res. 2003;92:809–16.PubMedCrossRef
20.
Zurück zum Zitat Masuo M, Reardon S, Ikebe M, Kitazawa T. A novel mechanism for the Ca2+-sensitizing effect of protein kinase C on vascular smooth muscle: inhibition of myosin light chain phosphatase. J Gen Physiol. 1994;104(2):265–86.PubMedCrossRef Masuo M, Reardon S, Ikebe M, Kitazawa T. A novel mechanism for the Ca2+-sensitizing effect of protein kinase C on vascular smooth muscle: inhibition of myosin light chain phosphatase. J Gen Physiol. 1994;104(2):265–86.PubMedCrossRef
21.
Zurück zum Zitat Nishizaw S, Neuz N, Uemura K. Direct evidence for a key role of protein kinase C in the development of vasospasm after subarachnoid hemorrhage. J Neurosurg. 1992;76:635–9.CrossRef Nishizaw S, Neuz N, Uemura K. Direct evidence for a key role of protein kinase C in the development of vasospasm after subarachnoid hemorrhage. J Neurosurg. 1992;76:635–9.CrossRef
22.
Zurück zum Zitat Nishizawa S, Yamamoto S, Yokoyama T, Ryu H, Uemura K. Chronological changes of arterial diameter, cGMP, and protein kinase C in the development of vasospasm. Stroke. 1995;26:1916–21.PubMedCrossRef Nishizawa S, Yamamoto S, Yokoyama T, Ryu H, Uemura K. Chronological changes of arterial diameter, cGMP, and protein kinase C in the development of vasospasm. Stroke. 1995;26:1916–21.PubMedCrossRef
23.
Zurück zum Zitat Joshi CN, Martin DN, Shaver P, Madamanchi C, Muller-Borer BJ, Tulis DA. Control of vascular smooth muscle cell growth by connexin 43. Front Physiol. 2012;3:220.PubMedPubMedCentralCrossRef Joshi CN, Martin DN, Shaver P, Madamanchi C, Muller-Borer BJ, Tulis DA. Control of vascular smooth muscle cell growth by connexin 43. Front Physiol. 2012;3:220.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Nishizawa S, Obara K, Nakayama K, Koide M, Yokoyama T, Yokota N, Ohta S. Protein kinase Cδ and α are involved in the development of vasospasm after subarachnoid hemorrhage. Eur J Pharmacol. 2000;398:113–9.PubMedCrossRef Nishizawa S, Obara K, Nakayama K, Koide M, Yokoyama T, Yokota N, Ohta S. Protein kinase Cδ and α are involved in the development of vasospasm after subarachnoid hemorrhage. Eur J Pharmacol. 2000;398:113–9.PubMedCrossRef
25.
Zurück zum Zitat Guan YY, Weir BK, Marton LS, Macdonald RL, Zhang H. Effects of erythrocyte lysate of different incubation times on intracellular free calcium in rat basilar artery smooth-muscle cells. J Neurosurg. 1998;89(6):1007–14.PubMedCrossRef Guan YY, Weir BK, Marton LS, Macdonald RL, Zhang H. Effects of erythrocyte lysate of different incubation times on intracellular free calcium in rat basilar artery smooth-muscle cells. J Neurosurg. 1998;89(6):1007–14.PubMedCrossRef
26.
Zurück zum Zitat Ishiguro M, Morielli AD, Zvarova K, Tranmer BI, Penar PL, Wellman GC. Oxyhemoglobin-induced suppression of voltage-dependent K+ channels in cerebral arteries by enhanced tyrosine kinase activity. Circ Res. 2006;99(11):1252–60.PubMedCrossRef Ishiguro M, Morielli AD, Zvarova K, Tranmer BI, Penar PL, Wellman GC. Oxyhemoglobin-induced suppression of voltage-dependent K+ channels in cerebral arteries by enhanced tyrosine kinase activity. Circ Res. 2006;99(11):1252–60.PubMedCrossRef
27.
Zurück zum Zitat Herbert JM, Augereau JM, Gleye J, Maffrand JP. Chelerythrine is a potent and specific inhibitor of protein kinase C. Biochem Biophys Res Commun. 1990;172(3):993–9.PubMedCrossRef Herbert JM, Augereau JM, Gleye J, Maffrand JP. Chelerythrine is a potent and specific inhibitor of protein kinase C. Biochem Biophys Res Commun. 1990;172(3):993–9.PubMedCrossRef
28.
Zurück zum Zitat Toullec D, Pianetti P, Coste H, Bellevergue P, Grand-Perret T, Ajakane M, et al. The bisindolylmaleimide GF 109203X is a potent and selective inhibitor of protein kinase C. J Biol Chem. 1991;266(24):15771–81.PubMed Toullec D, Pianetti P, Coste H, Bellevergue P, Grand-Perret T, Ajakane M, et al. The bisindolylmaleimide GF 109203X is a potent and selective inhibitor of protein kinase C. J Biol Chem. 1991;266(24):15771–81.PubMed
29.
Zurück zum Zitat O’Donnell JJ, Birukova AA, Beyer EC, Birukov KG. Gap junction protein connexin43 exacerbates lung vascular permeability. PLoS ONE. 2014;9(6):e100931.PubMedPubMedCentralCrossRef O’Donnell JJ, Birukova AA, Beyer EC, Birukov KG. Gap junction protein connexin43 exacerbates lung vascular permeability. PLoS ONE. 2014;9(6):e100931.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Gemel J, Nelson TK, Burt JM, Beyer EC. Inducible coexpression of connexin37 or connexin40 with connexin43 selectively affects intercellular molecular transfer. J Membr Biol. 2012;245(5–6):231–41.PubMedPubMedCentralCrossRef Gemel J, Nelson TK, Burt JM, Beyer EC. Inducible coexpression of connexin37 or connexin40 with connexin43 selectively affects intercellular molecular transfer. J Membr Biol. 2012;245(5–6):231–41.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Yang L, Lai WT, Wu YS, Zhang JA, Zhou XH, Yan J, et al. Simple and efficient rat model for studying delayed cerebral ischemia after subarachnoid hemorrhage. J Neurosci Methods. 2018;304:146–53.PubMedCrossRef Yang L, Lai WT, Wu YS, Zhang JA, Zhou XH, Yan J, et al. Simple and efficient rat model for studying delayed cerebral ischemia after subarachnoid hemorrhage. J Neurosci Methods. 2018;304:146–53.PubMedCrossRef
32.
Zurück zum Zitat Kaneda Y, Nakajima T, Nishikawa T, Yamamoto S, Ikegami H, Suzuki N, et al. Hemagglutinating virus of Japan (HVJ) envelope vector as a versatile gene delivery system. Mol Ther. 2002;6(2):219–26.PubMedCrossRef Kaneda Y, Nakajima T, Nishikawa T, Yamamoto S, Ikegami H, Suzuki N, et al. Hemagglutinating virus of Japan (HVJ) envelope vector as a versatile gene delivery system. Mol Ther. 2002;6(2):219–26.PubMedCrossRef
33.
Zurück zum Zitat Morita K, Motoyama N, Kitayama T, Morioka N, Kifune K, Dohi T. Spinal antiallodynia action of glycine transporter inhibitors in neuropathic pain models in mice. J Pharmacol Exp Ther. 2008;326(2):633–45.PubMedCrossRef Morita K, Motoyama N, Kitayama T, Morioka N, Kifune K, Dohi T. Spinal antiallodynia action of glycine transporter inhibitors in neuropathic pain models in mice. J Pharmacol Exp Ther. 2008;326(2):633–45.PubMedCrossRef
34.
Zurück zum Zitat Gurbi B, Brauswetter D, Varga A, Gyulavári P, Pénzes K, Murányi J, et al. The potential impact of connexin 43 expression on Bcl-2 protein level and taxane sensitivity in head and neck cancers—in vitro studies. Cancers. 2019;11(12):1848.CrossRefPubMedCentral Gurbi B, Brauswetter D, Varga A, Gyulavári P, Pénzes K, Murányi J, et al. The potential impact of connexin 43 expression on Bcl-2 protein level and taxane sensitivity in head and neck cancers—in vitro studies. Cancers. 2019;11(12):1848.CrossRefPubMedCentral
35.
Zurück zum Zitat Debarba LK, Vechiato F, Veida-Silva H, Borges BC, Jamur MC, Antunes-Rodrigues J, et al. The role of TCPTP on leptin effects on astrocyte morphology. Mol Cell Endocrinol. 2019;482:62–9.PubMedCrossRef Debarba LK, Vechiato F, Veida-Silva H, Borges BC, Jamur MC, Antunes-Rodrigues J, et al. The role of TCPTP on leptin effects on astrocyte morphology. Mol Cell Endocrinol. 2019;482:62–9.PubMedCrossRef
36.
Zurück zum Zitat Alexandrov AV, Sloan MA, Tegeler CH, Newell DN, Lumsden A, Garami Z, et al. Practice standards for transcranial Doppler (TCD) ultrasound. Part II. Clinical indications and expected outcomes. J Neuroimaging. 2012;22(3):215–24.PubMedCrossRef Alexandrov AV, Sloan MA, Tegeler CH, Newell DN, Lumsden A, Garami Z, et al. Practice standards for transcranial Doppler (TCD) ultrasound. Part II. Clinical indications and expected outcomes. J Neuroimaging. 2012;22(3):215–24.PubMedCrossRef
37.
Zurück zum Zitat Güresir E, Vasiliadis N, Dias S, Raab P, Seifert V, Vatter H. The effect of common carotid artery occlusion on delayed brain tissue damage in the rat double subarachnoid hemorrhage model. Acta Neurochir (Wien). 2012;154(1):11–9.PubMedCrossRef Güresir E, Vasiliadis N, Dias S, Raab P, Seifert V, Vatter H. The effect of common carotid artery occlusion on delayed brain tissue damage in the rat double subarachnoid hemorrhage model. Acta Neurochir (Wien). 2012;154(1):11–9.PubMedCrossRef
38.
Zurück zum Zitat Friedrich B, Müller F, Feiler S, Schöller K, Plesnila N. Experimental subarachnoid hemorrhage causes early and long-lasting microarterial constriction and microthrombosis: an in vivo microscopy study. J Cereb Blood Flow Metab. 2012;32(3):447–55.PubMedCrossRef Friedrich B, Müller F, Feiler S, Schöller K, Plesnila N. Experimental subarachnoid hemorrhage causes early and long-lasting microarterial constriction and microthrombosis: an in vivo microscopy study. J Cereb Blood Flow Metab. 2012;32(3):447–55.PubMedCrossRef
39.
Zurück zum Zitat Kataoka H, Kim SW, Plesnila N. Leukocyte–endothelium interactions during permanent focal cerebral ischemia in mice. J Cereb Blood Flow Metab. 2004;24(6):668–76.PubMedCrossRef Kataoka H, Kim SW, Plesnila N. Leukocyte–endothelium interactions during permanent focal cerebral ischemia in mice. J Cereb Blood Flow Metab. 2004;24(6):668–76.PubMedCrossRef
40.
Zurück zum Zitat Schwarzmaier SM, Kim SW, Trabold R, Plesnila N. Temporal profile of thrombogenesis in the cerebral microcirculation after traumatic brain injury in mice. J Neurotrauma. 2010;27(1):121–30.PubMedCrossRef Schwarzmaier SM, Kim SW, Trabold R, Plesnila N. Temporal profile of thrombogenesis in the cerebral microcirculation after traumatic brain injury in mice. J Neurotrauma. 2010;27(1):121–30.PubMedCrossRef
41.
Zurück zum Zitat Gabriels JE, Paul DL. Connexin43 is highly localized to sites of disturbed flow in rat aortic endothelium but connexin37 and connexin40 are more uniformly distributed. Circ Res. 1998;83(6):636–43.PubMedCrossRef Gabriels JE, Paul DL. Connexin43 is highly localized to sites of disturbed flow in rat aortic endothelium but connexin37 and connexin40 are more uniformly distributed. Circ Res. 1998;83(6):636–43.PubMedCrossRef
42.
Zurück zum Zitat Beck J, Raabe A, Lanfermann H, Seifert V, Weidauer S. Tissue at risk concept for endovascular treatment of severe vasospasm after aneurysmal subarachnoid haemorrhage. J Neurol Neurosurg Psychiatry. 2004;75(12):1779–81.PubMedPubMedCentralCrossRef Beck J, Raabe A, Lanfermann H, Seifert V, Weidauer S. Tissue at risk concept for endovascular treatment of severe vasospasm after aneurysmal subarachnoid haemorrhage. J Neurol Neurosurg Psychiatry. 2004;75(12):1779–81.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Jiang Q, Zhang RL, Zhang ZG, Ewing JR, Divine GW, Chopp M. Diffusion-, T2-, and perfusion-weighted nuclear magnetic resonance imaging of middle cerebral artery embolic stroke and recombinant tissue plasminogen activator intervention in the rat. J Cereb Blood Flow Metab. 1998;18(7):758–67.PubMedCrossRef Jiang Q, Zhang RL, Zhang ZG, Ewing JR, Divine GW, Chopp M. Diffusion-, T2-, and perfusion-weighted nuclear magnetic resonance imaging of middle cerebral artery embolic stroke and recombinant tissue plasminogen activator intervention in the rat. J Cereb Blood Flow Metab. 1998;18(7):758–67.PubMedCrossRef
44.
Zurück zum Zitat Leclerc X, Fichten A, Gauvrit JY, Riegel B, Steinling M, Lejeune JP, et al. Symptomatic vasospasm after subarachnoid haemorrhage: assessment of brain damage by diffusion and perfusion-weighted MRI and single-photon emission computed tomography. Neuroradiology. 2002;44(7):610–6.PubMedCrossRef Leclerc X, Fichten A, Gauvrit JY, Riegel B, Steinling M, Lejeune JP, et al. Symptomatic vasospasm after subarachnoid haemorrhage: assessment of brain damage by diffusion and perfusion-weighted MRI and single-photon emission computed tomography. Neuroradiology. 2002;44(7):610–6.PubMedCrossRef
45.
Zurück zum Zitat Solomon RA, Antunes JL, Chen RY, Bland L, Chien S. Decrease in cerebral blood flow in rats after experimental subarachnoid hemorrhage: a new animal model. Stroke. 1985;16(1):58–64.PubMedCrossRef Solomon RA, Antunes JL, Chen RY, Bland L, Chien S. Decrease in cerebral blood flow in rats after experimental subarachnoid hemorrhage: a new animal model. Stroke. 1985;16(1):58–64.PubMedCrossRef
46.
Zurück zum Zitat Solomon RA, Lovitz RL, Hegemann MT, Schuessler GB, Young WL, Chien S. Regional cerebral metabolic activity in the rat following experimental subarachnoid hemorrhage. J Cereb Blood Flow Metab. 1987;7(2):193–8.PubMedCrossRef Solomon RA, Lovitz RL, Hegemann MT, Schuessler GB, Young WL, Chien S. Regional cerebral metabolic activity in the rat following experimental subarachnoid hemorrhage. J Cereb Blood Flow Metab. 1987;7(2):193–8.PubMedCrossRef
47.
Zurück zum Zitat Granstam E, Granstam SO. Involvement of nitric oxide in the regulation of regional hemodynamics in streptozotocin-diabetic rats. Physiol Res. 2003;52(2):159.PubMed Granstam E, Granstam SO. Involvement of nitric oxide in the regulation of regional hemodynamics in streptozotocin-diabetic rats. Physiol Res. 2003;52(2):159.PubMed
48.
Zurück zum Zitat Palmer GM, Cairns BE, Berkes SL, Dunning PS, Taylor GA, Berde CB. The effects of lidocaine and adrenergic agonists on rat sciatic nerve and skeletal muscle blood flow in vivo. Anesth Analg. 2002;95(4):1080–6.PubMed Palmer GM, Cairns BE, Berkes SL, Dunning PS, Taylor GA, Berde CB. The effects of lidocaine and adrenergic agonists on rat sciatic nerve and skeletal muscle blood flow in vivo. Anesth Analg. 2002;95(4):1080–6.PubMed
49.
Zurück zum Zitat Wang H, Hong T, Wang H, Wang Y. Altered expression of connexin43 and its possible role in endothelin-1-induced contraction in rabbit basilar artery. Neurol Res. 2009;31(1):67–73.PubMedCrossRef Wang H, Hong T, Wang H, Wang Y. Altered expression of connexin43 and its possible role in endothelin-1-induced contraction in rabbit basilar artery. Neurol Res. 2009;31(1):67–73.PubMedCrossRef
50.
Zurück zum Zitat Sossin WS. Isoform specificity of protein kinase Cs in synaptic plasticity. Learn Mem. 2007;14(4):236–46.PubMedCrossRef Sossin WS. Isoform specificity of protein kinase Cs in synaptic plasticity. Learn Mem. 2007;14(4):236–46.PubMedCrossRef
51.
Zurück zum Zitat Richards TS, Dunn CA, Carter WG, Usui ML, Olerud JE, Lampe PD. Protein kinase C spatially and temporally regulates gap junctional communication during human wound repair via phosphorylation of connexin43 on serine368. J Cell Biol. 2004;167(3):555–62.PubMedPubMedCentralCrossRef Richards TS, Dunn CA, Carter WG, Usui ML, Olerud JE, Lampe PD. Protein kinase C spatially and temporally regulates gap junctional communication during human wound repair via phosphorylation of connexin43 on serine368. J Cell Biol. 2004;167(3):555–62.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Owada-Makabe K, Tsubota Y, Yukawa K, Kakimoto N, Liang XM, Ichinose M, et al. Direct in vivo protein transduction into a specific restricted brain area in rats. Neurosci Lett. 2005;378(1):18–21.PubMedCrossRef Owada-Makabe K, Tsubota Y, Yukawa K, Kakimoto N, Liang XM, Ichinose M, et al. Direct in vivo protein transduction into a specific restricted brain area in rats. Neurosci Lett. 2005;378(1):18–21.PubMedCrossRef
53.
Zurück zum Zitat Yuki S, Kondo Y, Kato F, Kato M, Matsuo N. Noncytotoxic ribonuclease, RNase T1, induces tumor cell death via hemagglutinating virus of Japan envelope vector. Eur J Biochem. 2004;271(17):3567–72.PubMedCrossRef Yuki S, Kondo Y, Kato F, Kato M, Matsuo N. Noncytotoxic ribonuclease, RNase T1, induces tumor cell death via hemagglutinating virus of Japan envelope vector. Eur J Biochem. 2004;271(17):3567–72.PubMedCrossRef
54.
Zurück zum Zitat Ohkuma H, Itoh K, Shibata S, Suzuki S. Morphological changes of intraparenchymal arterioles after experimental subarachnoid hemorrhage in dogs. Neurosurgery. 1997;41(1):230–6 Discussion 235–6.PubMedCrossRef Ohkuma H, Itoh K, Shibata S, Suzuki S. Morphological changes of intraparenchymal arterioles after experimental subarachnoid hemorrhage in dogs. Neurosurgery. 1997;41(1):230–6 Discussion 235–6.PubMedCrossRef
55.
Zurück zum Zitat Uhl E, Lehmberg J, Steiger HJ, Messmer K. Intraoperative detection of early microvasospasm in patients with subarachnoid hemorrhage by using orthogonal polarization spectral imaging. Neurosurgery. 2003;52(6):1307–17 Discussion 1315–7.PubMedCrossRef Uhl E, Lehmberg J, Steiger HJ, Messmer K. Intraoperative detection of early microvasospasm in patients with subarachnoid hemorrhage by using orthogonal polarization spectral imaging. Neurosurgery. 2003;52(6):1307–17 Discussion 1315–7.PubMedCrossRef
56.
Zurück zum Zitat Sun BL, Zheng CB, Yang MF, Yuan H, Zhang SM, Wang LX. Dynamic alterations of cerebral pial microcirculation during experimental subarachnoid hemorrhage. Cell Mol Neurobiol. 2009;29(2):235–41.PubMedCrossRef Sun BL, Zheng CB, Yang MF, Yuan H, Zhang SM, Wang LX. Dynamic alterations of cerebral pial microcirculation during experimental subarachnoid hemorrhage. Cell Mol Neurobiol. 2009;29(2):235–41.PubMedCrossRef
57.
Zurück zum Zitat Ungvari Z, Csiszar A, Koller A. Increases in endothelial Ca2+ activate K(Ca) channels and elicit EDHF-type arteriolar dilation via gap junctions. Am J Physiol Heart Circ Physiol. 2002;282(5):H1760–7.PubMedCrossRef Ungvari Z, Csiszar A, Koller A. Increases in endothelial Ca2+ activate K(Ca) channels and elicit EDHF-type arteriolar dilation via gap junctions. Am J Physiol Heart Circ Physiol. 2002;282(5):H1760–7.PubMedCrossRef
58.
Zurück zum Zitat Bagher P, Segal SS. Regulation of blood flow in the microcirculation: role of conducted vasodilation. Acta Physiol (Oxf). 2011;202(3):271–84.PubMedCentralCrossRef Bagher P, Segal SS. Regulation of blood flow in the microcirculation: role of conducted vasodilation. Acta Physiol (Oxf). 2011;202(3):271–84.PubMedCentralCrossRef
59.
Zurück zum Zitat Hoepfl B, Rodenwaldt B, Pohl U, De Wit C. EDHF, but not NO or prostaglandins, is critical to evoke a conducted dilation upon ACh in hamster arterioles. Am J Physiol Heart Circ Physiol. 2002;283(3):H996–1004.PubMedCrossRef Hoepfl B, Rodenwaldt B, Pohl U, De Wit C. EDHF, but not NO or prostaglandins, is critical to evoke a conducted dilation upon ACh in hamster arterioles. Am J Physiol Heart Circ Physiol. 2002;283(3):H996–1004.PubMedCrossRef
60.
Zurück zum Zitat Déglise S, Martin D, Probst H, Saucy F, Hayoz D, Waeber G, et al. Increased connexin43 expression in human saphenous veins in culture is associated with intimal hyperplasia. J Vasc Surg. 2005;41(6):1043–52.PubMedCrossRef Déglise S, Martin D, Probst H, Saucy F, Hayoz D, Waeber G, et al. Increased connexin43 expression in human saphenous veins in culture is associated with intimal hyperplasia. J Vasc Surg. 2005;41(6):1043–52.PubMedCrossRef
61.
Zurück zum Zitat Chaytor AT, Bakker LM, Edwards DH, Griffith TM. Connexin-mimetic peptides dissociate electrotonic EDHF-type signalling via myoendothelial and smooth muscle gap junctions in the rabbit iliac artery. Br J Pharmacol. 2005;144(1):108–14.PubMedCrossRef Chaytor AT, Bakker LM, Edwards DH, Griffith TM. Connexin-mimetic peptides dissociate electrotonic EDHF-type signalling via myoendothelial and smooth muscle gap junctions in the rabbit iliac artery. Br J Pharmacol. 2005;144(1):108–14.PubMedCrossRef
62.
Zurück zum Zitat Little TL, Beyer EC, Duling BR. Connexin 43 and connexin 40 gap junctional proteins are present in arteriolar smooth muscle and endothelium in vivo. Am J Physiol. 1995;268(2 Pt 2):H729–39.PubMed Little TL, Beyer EC, Duling BR. Connexin 43 and connexin 40 gap junctional proteins are present in arteriolar smooth muscle and endothelium in vivo. Am J Physiol. 1995;268(2 Pt 2):H729–39.PubMed
63.
Zurück zum Zitat van Kempen MJ, Jongsma HJ. Distribution of connexin37, connexin40 and connexin43 in the aorta and coronary artery of several mammals. Histochem Cell Biol. 1999;112(6):479–86.PubMedCrossRef van Kempen MJ, Jongsma HJ. Distribution of connexin37, connexin40 and connexin43 in the aorta and coronary artery of several mammals. Histochem Cell Biol. 1999;112(6):479–86.PubMedCrossRef
64.
Zurück zum Zitat de Wit C, Roos F, Bolz SS, Kirchhoff S, Krüger O, Willecke K, et al. Impaired conduction of vasodilation along arterioles in connexin40-deficient mice. Circ Res. 2000;86(6):649–55.PubMedCrossRef de Wit C, Roos F, Bolz SS, Kirchhoff S, Krüger O, Willecke K, et al. Impaired conduction of vasodilation along arterioles in connexin40-deficient mice. Circ Res. 2000;86(6):649–55.PubMedCrossRef
65.
Zurück zum Zitat de Wit C, Roos F, Bolz SS, Pohl U. Lack of vascular connexin 40 is associated with hypertension and irregular arteriolar vasomotion. Physiol Genomics. 2003;13(2):169–77.PubMedCrossRef de Wit C, Roos F, Bolz SS, Pohl U. Lack of vascular connexin 40 is associated with hypertension and irregular arteriolar vasomotion. Physiol Genomics. 2003;13(2):169–77.PubMedCrossRef
66.
Zurück zum Zitat Mather S, Dora KA, Sandow SL, Winter P, Garland CJ. Rapid endothelial cell-selective loading of connexin 40 antibody blocks endothelium-derived hyperpolarizing factor dilation in rat small mesenteric arteries. Circ Res. 2005;97(4):399–407.PubMedCrossRef Mather S, Dora KA, Sandow SL, Winter P, Garland CJ. Rapid endothelial cell-selective loading of connexin 40 antibody blocks endothelium-derived hyperpolarizing factor dilation in rat small mesenteric arteries. Circ Res. 2005;97(4):399–407.PubMedCrossRef
67.
Zurück zum Zitat Burt JM, Fletcher AM, Steele TD, Wu Y, Cottrell GT, Kurjiaka DT. Alteration of Cx43:Cx40 expression ratio in A7r5 cells. Am J Physiol Cell Physiol. 2001;280(3):C500–8.PubMedCrossRef Burt JM, Fletcher AM, Steele TD, Wu Y, Cottrell GT, Kurjiaka DT. Alteration of Cx43:Cx40 expression ratio in A7r5 cells. Am J Physiol Cell Physiol. 2001;280(3):C500–8.PubMedCrossRef
Metadaten
Titel
The important role of connexin 43 in subarachnoid hemorrhage-induced cerebral vasospasm
verfasst von
Le Yang
Jian Yan
Jin-An Zhang
Xin-Hui Zhou
Chao Fang
Er-Ming Zeng
Bin Tang
Jian Duan
Guo-Hui Lu
Tao Hong
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2019
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-019-02190-1

Weitere Artikel der Ausgabe 1/2019

Journal of Translational Medicine 1/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.