Introduction
Myocardial infarction (MI) shows the characteristics of autonomic nerve imbalance with an increased ratio of sympathetic nerve to vagus nerve activity, accompanied by an inflammatory response and recruitment of leukocytes, leading to myocardial injury.1 Basic and clinical studies have shown that vagal nerve stimulation (VNS) balances autonomic nerve activity in injured hearts, providing a novel therapeutic strategy for autonomic dysfunction by activating cholinergic anti-inflammatory pathways.1–4 However, whether VNS-mediated anti-inflammatory roles are beneficial to angiogenesis of the infarcted heart is still unclear.
Angiogenesis is essential to the surviving myocardium and the recovery of ventricular function after MI by reshaping the blood supply to the heart.5 The reduced levels of α7-nicotinic acetylcholine (ACh) receptor (α7-nAChR) expression hamper angiogenesis in ischaemic or infarcted hearts.6,7 Indeed, coinciding with vagal innervations in the conduction system and coronary vessels of the heart,8 VNS increased the concentration of ACh in the ventricle.9–11 Meanwhile, VNS restored and even enhanced the levels of α7-nAChR and m3-AChR expression in the injured heart,12 indicating that VNS could have beneficial involvement in angiogenesis of infarcted heart through released ACh and its activated α7-nAChR and/or m3-AChR. Unfortunately, its potential mechanism has not been fully explored.
Our and other scholars' results have shown that vascular endothelial growth factor (VEGF) is involved in the angiogenesis of ischaemic or infarcted hearts.12–14 Of interest, VEGF-A gene therapy alone does not effectively result in sufficient angiogenesis to match the improvement in cardiac function,15 implying that other underlying factors may also be required for angiogenesis in injured hearts. Indeed, in addition to typically regulating leukocyte trafficking, recent evidence has shown that chemokines are cardioprotective and participate in angiogenesis.16-18 As a classical chemokine molecule, stromal cell-derived factor-l alpha (SDF-1α) can activate and/or induce the migration of haematopoietic/endothelial progenitor cells and endothelial cells, in addition to most leukocytes.19–22 However, whether SDF-1α is involved in VNS-mediated anti-inflammatory effects and angiogenesis has yet to be determined.
In this study, rats with MI were treated with VNS, and increased SDF-1α expression and CD31-positive vessel density in the infarcted heart were found, which could be abolished by the m/nAChR inhibitors mecamylamine and atropine or knockdown of SDF-1α by shRNA. In vitro, ACh induced SDF-1α expression and its redistribution along the branch of the formed tube in human coronary artery endothelial cells (HCAECs), which could be related to the m/nAChR-AKT-Sp1 signalling pathway. Taken together, these results indicate that vagus nerve stimulation (VNS)-induced SDF-1α promotes angiogenesis and repair in infarcted hearts through the m/nAChR-AKT-Sp1 signalling pathway.
Methods
Animals
In accordance with the Guide for the Care and Use of Laboratory Animals published by China and US National Institutes of Health, adult male Sprague–Dawley (SD) rats (250–300 g) were supplied by the Experimental Animal Centre of the Hubei University of Medicine. All animal protocols were approved by the Institutional Animal Care and Use Committee of Hubei University of Medicine (HBUM2018-142).
Myocardial infarction model establishment
The left anterior descending coronary artery in rats was ligated to prepare the MI model as previously described.23 In brief, ketamine (50 mg/kg, i.p.) and xylazine (10 mg/kg, i.p.) were used to anaesthetize SD rats (250–300 g). Descending coronary artery ligation was promptly performed after thoracotomy on the left fourth intercostal space after tracheal ventilation remained stable using a Colombus ventilator (HX-300, Taimeng Instruments, Chengdu, China). When confirming the occurrence of MI with blanching of the myocardium as well as electrocardiography, the open thoracic cavity was sutured layer by layer immediately.
Method of vagus nerve stimulation
Surviving rats were randomized into groups with sham or stimulation on the seventh day of MI. After the left vagal nerve in the neck was gently exposed and separated, the vagal nerve was looped with Teflon-coated stainless-steel wires (UL1330; Triumph Cable Co, Ltd, China) and ligated into the stimulator output part (BL-420S; Chengdu Tme Technology Co, Ltd, China) for electrical stimulation (20 Hz for 10 s every minute for 4 h) as previously described.13 Regular pulse stimulation in the vagal nerve was executed in the VNS group.13 Similar operations were performed without initiating stimulation in the sham group (MI). A 10% reduction in heart rate was used as a criterion for VNS. A mixture of white petrolatum (Vaseline) and paraffin was used to immerse the vagus nerve and connected electrodes to prevent drying.
Acetylcholine receptor inhibitor
One hour before VNS, mecamylamine (10 mg/kg, ip) or atropine (10 mg/kg, ip) was used to assess whether the role of VNS in SDF-1α expression in the infarcted heart was linked to mACh-R and α7-nAChR, as described previously.12,13
Knockdown of stromal cell-derived factor-l alpha by shRNA
SDF-1α shRNA (shSDF-1) was designed using a dedicated program provided by our published data.15 To determine the effect of SDF-1α on angiogenesis in the infarcted heart, Ad-shCtrl and Ad-shSDF-1α (1 × 109 pfu in 200 μL) were injected into four sites of the infracted hearts (50 μL per site, 12 rats per group) with a 30-gauge tuberculin syringe 3 days before VNS. Two injections were in the myocardium bordering the ischaemic area, and two were within the ischaemic area. Penicillin (150 000 U/mL, i.v.) was given before each procedure. Buprenorphine hydrochloride (0.05 mg/kg, s.c.) was administered twice a day for the first 48 h after the procedure.
Measurement of coronary flow and haemodynamic parameters
Six SD rats (250–300 g)/group were anaesthetized with ketamine (50 mg/kg, i.p.) and xylazine (10 mg/kg, i.p.). A thoracotomy was performed, and hearts were rapidly excised into ice-cold perfusion fluid. The aorta was cannulated on a shortened and blunted 14-gauge needle, and perfusion initiated at a constant pressure of 80 mmHg on the Langendorff system. A fluid-filled balloon constructed from polyvinyl chloride film was introduced into the left ventricle through an incision in the left atrial appendage. Two tubes were introduced into the right atrial appendage through the superior and inferior caval veins. Hearts were immersed in warmed perfusate in a jacketed bath maintained at 37°C, and perfusate delivered to the coronary circulation was maintained at the same temperature using a super constant temperature water bath. The organ bath and perfusate temperatures were continuously monitored using a digital thermometer. Langendorff-perfused hearts were allowed to equilibrate until heart rate and contractility reached steady state for 30 min. Total coronary flow within 30 min was collected in the cylinder through tubes linked to the right atrial appendage for evaluating coronary flow percentage/min. Krebs–Henseleit perfusion fluid (containing NaCl, 119 mM; glucose, 11 mM; NaHCO3, 22 mM; KCl, 4.7 mM; MgCl2, 1.2 mM; KH2PO4, 1.2 mM; CaCl2, 2.5 mM; EDTA, 0.5 mM; and pyruvate, 2 mM) was used in all experiments. The fluid with a pH of 7.4 was bubbled with a mixture of 95% O2 and 5% CO2 at 37°C and was filtered through a 0.45 μm filter before delivery to the heart.14
Twenty-eight days after VNS treatment, heart functions, including left ventricular systolic pressure, left ventricular end-diastolic pressure, and rate of the rise and fall of ventricular pressure (+dP/dtmax and –dP/dtmax), were evaluated as described previously.23 Following anaesthesia for rats with the application of ketamine (50 mg/kg, i.p.) and xylazine (10 mg/kg, i.p.), one end of the catheter was connected to a pressure transducer, and the other end of the catheter filled with heparinized (10 U/mL) saline solution was advanced into the left ventricle to record ventricular pressure while it was inserted into the isolated left carotid artery. After evaluating cardiac function, the heart was immediately collected for subsequent detection and analysis.
Heart tissue immunostaining
Serial transverse sections of heart tissue (5 μm) were prepared as previously described.15 Before adding primary antibodies, a blocking buffer (PBS containing 5% goat serum and 0.1% Triton X-100) was used to treat these sections at room temperature for 1 h. The primary antibodies (diluted in blocking buffer), including goat anti-rat SDF-1α (sc-6193, 1:150; Santa Cruz), mouse anti-rat CD31 (ab24590, 1:250; Abcam), rabbit anti-rat CD68 (1:250, GB11067, ServiceBio), and rabbit anti-rat peNOS (#9575, 1:250, CST), were incubated at 4°C overnight; then, the secondary antibodies, including horseradish peroxidase (HRP)-labelled goat anti-mouse IgG, goat-anti-rabbit IgG, FITC-conjugated anti-rabbit IgG, or TRITC-conjugated anti-mouse IgG (Jackson ImmunoResearch), were incubated at room temperature for 2 h.15 Eventually, these indicated results were quantified by densitometry analysis (Image Pro, USA) after taking pictures under a microscope (MF43-N, Olympus, Japan).
Establishment of an inflammatory injury model in human coronary artery endothelial cells induced by lipopolysaccharide
HCAECs (Jennio Biotech Co., Ltd., Guangzhou, China) were cultured in complete medium for 24 h as previously mentioned.14 Then, lipopolysaccharide (LPS, 1 mg/μL) was added to the medium for 24 h. Successfully embellishment of the LPS-induced HCAEC inflammatory injury model in vitro was confirmed by evaluation of HCAEC-mediated tube formation and inflammatory responses. Evaluation of tube formation was shown in the HCAEC tube formation method. Inflammatory responses were assessed by detecting NF-κB p65 by using immunofluorescence staining and tumour necrosis factor α (TNF-α), interleukin 1β (IL-1β), and interleukin 6 (IL-6) by using ELISA.24,25
Human coronary artery endothelial cells tube formation
HCAEC tube formation was carried out as described,26 and HCAECs (4 × 105/mL) were cultured in 24-well plates coated with Matrigel™ for 12 h. To confirm the relationship between ACh and LPS during angiogenesis, ACh was added to the tube formation medium with or without LPS. To further confirm whether the role of ACh in HCAEC tube formation is associated with mACh-R and α7-nAChR, atropine (1 μM) or mecamylamine (7 μM) was used for 30 min before changing the tube formation medium. To ensure the reliability of the results, three independent experiments were performed. Three duplicated wells/groups were executed every time. The total number of tube branches (in pixels) per well was quantified using the Image-Pro Plus software package (Media Cybernetics, Carlsbad, CA) and averaged. The cells were finally fixed in formalin, washed with PBS, and imaged using microscopy.
Processing method of distribution characteristics of stromal cell derived factor 1 alpha/C-X-C motif chemokine receptor 4/7 in human coronary artery endothelial cells
The processing method of the distribution characteristics of SDF-1α/C-X-C motif chemokine receptor 4/7 (CXCR4/7) in HCAECs precultured in Matrigel was as follows: After HCAECs were cultured with complete medium in 6-well plates coated with Matrigel™ for 24 h, the cells were digested, collected and inoculated into 24-well plates with tube formation medium for 12 h. To confirm the relationship between ACh and LPS during angiogenesis, ACh was added to the tube formation medium with or without LPS. To further confirm whether the role of ACh in the expression and distribution characteristics of SDF-1α and CXCR4/7 is associated with mACh-R and α7-nAChR, atropine (1 μM) or mecamylamine (7 μM) was used for 30 min before changing the tube formation medium. The cells were finally fixed in formalin, washed with PBS, and subjected to immunofluorescence staining of SDF-1α/CXCR4/7. The cells were imaged to observe the expression and distribution characteristics of SDF-1α and CXCR4/7. Three independent experiments were performed.
The distribution characteristics of SDF-1α/CXCR4/7 in HCAECs were not precultured in Matrigel as follows: After HCAECs were cultured with complete medium in 6-well plates for 24 h, the cells were digested, collected and inoculated into 24-well plates with complete medium for 24 h. To confirm the relationship between ACh and LPS during angiogenesis, ACh was added to the tube formation medium with or without LPS. To further confirm whether the role of ACh in the expression and distribution characteristics of SDF-1α and CXCR4/7 is associated with mACh-R and α7-nAChR, atropine (1 μM) or mecamylamine (7 μM) was used for 30 min before changing the complete medium. The cells were finally fixed in formalin, washed with PBS, and subjected to immunofluorescence staining of SDF-1α/CXCR4/7. The cells were imaged to observe the distribution characteristics of SDF-1α and CXCR4/7. Three independent experiments were performed.
Immunofluorescence staining
HCAECs were incubated in blocking buffer (PBS containing 5% goat serum and 0.1% Triton X-100) at room temperature for 1 h. Incubation with antibodies (diluted 1:250 in blocking buffer) was performed at 4°C overnight for primary antibodies and at room temperature for 2 h for secondary antibodies. The primary antibodies used were mouse/rabbit anti-human SDF-1α (ab18919, 1:250; Abcam; sc-74271, 1:150, Santa Cruz), rabbit anti-human CXCR4 (sc-74271, NB100-74396, 1:300, NOVUS), rabbit anti-human CXCR7 (GB11256, 1:250; Servicebio), and WGA (L4895, 1:500, Sigma). The secondary antibodies were FITC-conjugated anti-rabbit IgG or TRITC-conjugated anti-mouse IgG (Jackson ImmunoResearch) (Tang et al., 2011; van Gils et al., 2013). Double staining of SDF-1α and CXCR4/7 was used to confirm the relationship of distribution between SDF-1α and CXCR4/7 during tube formation. Two rounds of wheat germ agglutinin (WGA-FITC) and SDF-1α/CXCR4/7 were used to confirm the indicated protein internalization and trafficking.27
Administration of inhibitors or adenovirus overexpression in human coronary artery endothelial cells
HCAECs (Jennio Biotech Co., Ltd., Guangzhou, China) were cultured in complete medium for 48 h. To further assess the mechanisms underlying ACh-induced SDF-1α expression in HCAECs, the mACh-R blocker atropine (1 μM) or the nACh-R blocker mecamylamine (7 μM), the PI3K/AKT blocker wortmannin (50 nM), the MEK/ERK1/2 inhibitor PD98059 (50 μM), the p38MAPK inhibitor SB203580 (30 μM) and the Sp1 blocker mithramycin (1 μM, Sp1-I) were inoculated for 1 h before treatment with 10−5 M ACh for 24 h. To further assess the role of Sp1 in ACh-induced SDF-1α expression in HCAECs, HCAECs were transfected with Ad-Sp1 at 100 MOI (Vigenebio, Shandong, China) for 24 h before treatment with 10−5 M ACh for 24 h. For subsequent target molecular detection, harvested cells were lysed in RIPA buffer containing protease and phosphatase inhibitors.
ELISA
ELISA for ACh was performed in cardiac tissue and serum treated with the cholinesterase inhibitor eserine (100 μM) after VNS using a commercial kit by following the manufacturer's protocol (ab65345, Abcam). ELISA for TNF-α (900-M73, PeproTech), IL-6 (900-M86, PeproTech) and IL-1β (900-M91, PeproTech) was performed in cardiac tissue and serum after VNS using a commercial kit by following the manufacturer's protocol. TNF-α (PT518, Beyotime, CN), IL-6 (PI330, Beyotime, CN) and IL-1β (PI305, Beyotime, CN) in the supernatant of HCAECs were detected by ELISA following the manufacturer's protocol.
Measurement of vascular density
The numbers of CD31-positive vessels in each section were analysed in 6 ~ 8 equally distributed areas of 0.1 mm2 in the peri-infarction or infarction area. The values were then expressed as the number of vessels per square millimetre. Two independent examiners analysed these results using the Image-Pro Plus software package (Media Cybernetics, Carlsbad, CA).28
Western blot
Western blotting was performed as previously described.29 Fifty micrograms of protein was resolved in a 10% SDS–PAGE gel and transferred onto a polyvinylidene fluoride membrane (Millipore). After blocking with 5% nonfat milk, the membrane was incubated overnight with primary antibodies, including rabbit anti-human SDF-1α (ab18919, 1:1000; Abcam), mouse anti-human SDF-1α (sc-74271, 1:500; Santa Cruz), Sp1 (sc-420, 1:500; Santa Cruz), rabbit anti-human p-Sp1 (ab37707, 1:1000; Abcam), rabbit anti-human V-akt murine thymoma viral oncogene homolog (AKT) (9272S, 1:500; Cell Signaling), rabbit anti-human phosphorylated AKT (pAKT) (9271S, 1:500; Cell Signaling), and mouse anti-human α-tubulin (YM3035, 1:500, Immunoway). Then, incubation with horseradish peroxidase (HRP)-conjugated secondary antibodies (anti-rabbit IgG, anti-mouse IgG, 1:10 000, Jackson ImmunoResearch) was carried out for 90 min. Eventually, the expression of the indicated proteins was quantified by densitometry analysis (Image Pro, USA) after visualization with an enhanced chemiluminescence reaction (Amersham Pharmacia Biotech).29
Transmission electron microscopy
Cardiac tissues were collected for transmission electron microscopic observation of endothelial cell morphology and its linkage and myocardial cell mitochondrial structure according to the manufacturer's protocol (Servicebio, CN).
Statistical analyses
Data shown are the mean ± SD. Statistical significance between two groups was determined by paired or unpaired Student's t test. The results for more than two experimental groups were evaluated by one-way ANOVA to specify differences between groups. P < 0.05 was considered significant.
Results
Vagus nerve stimulation promotes angiogenesis in the infarcted heart through m/n-ACh-R
To first identify whether VNS was successful in the rat MI model, ACh levels in heart tissues and serum were detected by ELISA, showing that ACh levels were increased after VNS (Figure 1A,B), indicating that VNS caused the cardiac vagus nerve to release more ACh. To further confirm whether VNS was successful in the rat MI model, TNF-α, IL-1β and IL-6 levels, as typical markers for the anti-inflammatory role of VNS in MI hearts, were analysed by ELISA, showing that TNF-α, IL-1β and IL-6 levels were obviously decreased in MI hearts treated with VNS (Figure 1C–E). These results demonstrated that VNS model establishment was successful for MI therapy.
[IMAGE OMITTED. SEE PDF]
Subsequently, to explore the role of VNS in angiogenesis in the infarcted heart, we used immunohistochemical staining for the endothelial cell marker CD31. As shown in Figure 1F,G, VNS increased the numbers of CD31-positive vessels in the infarction area and peri-infarction area of infarcted hearts, indicating that VNS induced angiogenesis in MI hearts. Meanwhile, VNS improved CD31-positive cell function characterized by increased eNOS levels (Figure 2A–C). Furthermore, transmission electron microscopy showed that VNS improved EC function with a relatively complete cell membrane and normal intercellular space (Figure 2D), resulting in the recovery of the morphology and structure of myocardial mitochondria adjacent to ECs (Figure 2E). These results demonstrated that successful VNS treatment promoted angiogenesis in the infarcted heart.
[IMAGE OMITTED. SEE PDF]
Finally, to further confirm whether VNS-induced angiogenesis involved cholinergic receptors, the m-AChR inhibitor atropine (0.5 mg/kg, ip) and the α7-nAChR mecamylamine (1.0 mg/kg, ip) were used 1 h before VNS. As shown in Figure 3A–C, atropine and mecamylamine obviously abolished the specific effects of VNS on angiogenesis (Figure 3A–D). These results demonstrated that VNS promoted angiogenesis in the infarcted heart through m/n-ACh-R.
[IMAGE OMITTED. SEE PDF]
Vagus nerve stimulation-induced angiogenesis involves stromal cell derived factor 1 alpha in the infarcted heart through m/n-ACh-R
Previous reports have shown that SDF-1α could play a crucial role in angiogenesis in the infarcted heart.30 We first found that VNS induced SDF-1α expression in CD31-positive cells, especially in epicardial endothelial cells (Figure 4A–C), leading to increased numbers of CD31-positive vessels in the infarcted hearts (Figure 3). Then, to confirm whether VNS-induced SDF-1α expression involved cholinergic receptors, the m-AChR inhibitor atropine (0.5 mg/kg, ip) and the α7-nAChR mecamylamine (1.0 mg/kg, ip) were used 1 h before VNS. As shown in Figure 4A–C, atropine and mecamylamine obviously abolished the specific effects of VNS on SDF-1α, indicating that m/n-AChR was involved in VNS-induced SDF-1α expression in coronary vessels. These results suggested that VNS-induced angiogenesis involves SDF-1α in the infarcted heart through m/n-ACh-R.
[IMAGE OMITTED. SEE PDF]
Stromal cell derived factor 1 alpha is involved in vagus nerve stimulation-induced angiogenesis in the infarcted heart
To further determine whether SDF-1α is involved in VNS-induced angiogenesis in the infarcted heart, local injection of Ad-shSDF-1α into the infarcted hearts was performed 3 days before MI. As shown in Figure 5A,B, application of Ad-shSDF-1α effectively knocked down SDF-1α expression in the infarcted hearts. Furthermore, with the knockdown of SDF-1α, we found that Ad-shSDF-1α significantly reduced the CD31-positive vascular density (Figure 5C,D). Functionally, the beneficial effects of VNS on coronary blood flow (Figure 5E) could be eliminated by the application of Ad-shSDF-1α. These data indicated that VNS-induced SDF-1α expression promoted angiogenesis through m/n-ACh-R.
[IMAGE OMITTED. SEE PDF]
Acetylcholine promoted human coronary artery endothelial cells tube formation while inhibiting the inflammatory response in vitro through m/n-AChR
To simulate the inflammatory reaction of MI, LPS (1 mg/μL) was added to the culture medium of HCAECs for 24 h. Nuclear translocation of NF-κB p65 and the levels of TNF-α, IL-1β and IL-6 were evaluated to identify whether the HCAEC inflammatory injury model was successfully established. As shown in Figure 6A,B, LPS promoted NF-κB p65 nuclear translocation in HCAECs, leading to an increase in TNF-α, IL-1β and IL-6 levels (Figure 6C–E). Meanwhile, LPS inhibited HCAEC tube formation in vitro (Figure 6F,G). These data indicated that the LPS-mediated HCAEC inflammatory injury model was successfully established.
[IMAGE OMITTED. SEE PDF]
Subsequently, to confirm whether VNS and the neurotransmitter ACh promoted angiogenesis, ACh was added to the HCAEC tube formation model in vitro to mimic the role of VNS in angiogenesis in vivo. As shown in Figure 6E,F, ACh in vitro completely reversed the inhibitory effect of LPS on tube formation, and the specific effect could be abolished by atropine or mecamylamine, indicating that ACh in vitro, similar to VNS in vivo, promoted angiogenesis in the inflammatory damage environment of MI through m/n-ACh-R.
Acetylcholine induced stromal cell derived factor 1 alpha distribution along new branches during tube formation
In addition to SDF-1α expression related to angiogenesis, immunofluorescence staining showed that SDF-1α was distributed along new branches of tube formation. Furthermore, ACh enhanced the specific distribution of SDF-1α during tube formation (Figure 7A,B). Under LPS stimulation, the typical characteristics of an SDF-1α distribution were altered, showing the distribution around the nucleus within round, conical, rod-shaped HCAECs. More importantly, ACh recovered the LPS-induced SDF-1α redistribution along new branches during the formation of the vascular ring (Figure 7A,B). These effects could be markedly cancelled by atropine or mecamylamine. These results suggested that ACh promoted angiogenesis by restoring SDF-1α distribution along new branches.
[IMAGE OMITTED. SEE PDF]
Acetylcholine induced C-X-C motif chemokine receptor 4 distribution along a parallel direction with stromal cell derived factor 1 alpha through m/n-AChR
To confirm the relationship between ACh and CXCR4 during HCAEC tube formation mediated by SDF-1α, double staining of SDF-1/CXCR4 by immunofluorescence showed CXCR4 distribution within normal HCAECs along a parallel direction with SDF-1α. LPS treatment altered CXCR4 distribution along the branch direction of tube formation; ACh could substantially eliminate the inhibitory role of LPS in CXCR4 distribution (Figure 8A,B). Furthermore, the ACh-mediated effect on CXCR4 distribution could be memorably abolished by atropine or mecamylamine. These results suggested that ACh recovers CXCR4 distribution in parallel with SDF-1α during tube formation through m/n-AChR.
[IMAGE OMITTED. SEE PDF]
CXCR7, another receptor for SDF-1α, showed changes similar to those of CXCR4 when facing LPS treatment with or without ACh, with the addition of atropine or mecamylamine (Figure S1).
Acetylcholine promoted stromal cell derived factor 1 alpha expression in human coronary artery endothelial cells through the AKT-Sp1 signalling pathway
To explore the possible molecular mechanism of VNS in SDF-1α expression, gene chip analysis was previously performed, showing that the PI3K-AKT signalling pathway was obviously altered in infarcted hearts following treatment with VNS and ACh.13 We first found that ACh dose-dependently increased the protein level of SDF-1α (Figure S2). Further findings showed that ACh did not alter the total AKT level but significantly increased the phosphorylated AKT level in HCAECs (Figure 9A,B). Moreover, the typical PI3K/AKT inhibitor wortmannin decreased ACh-mediated SDF-1α expression (Figure 7A,B). Meanwhile, ACh obviously enhanced the levels of p-Sp1 in HCAECs along with ACh-induced AKT phosphorylation (Figure 9C,D), and the specific role could almost be entirely blocked by atropine or mecamylamine (Figure 9C,D). Importantly, ACh-induced SDF-1α expression could almost entirely be abolished by the Sp1-specific inhibitor mithramycin A, while Sp1 overexpression enhanced ACh-induced SDF-1α expression (Figure 9E–H), indicating that ACh increased Sp1 phosphorylation and activity through pAKT. These results suggested that ACh-induced SDF-1α expression is involved in the m/nAChR/AKT/Sp1 signalling pathway.
[IMAGE OMITTED. SEE PDF]
Vagus nerve stimulation and acetylcholine improved cardiac function
Functional analysis was performed to explore whether VNS-induced angiogenesis improved infarcted heart function. The results showed that coronary blood flows and LV function, including left ventricular systolic pressure, left ventricular end-diastolic pressure, +dP/dtmax, and –dP/dtmax, was significantly improved in VNS-treated hearts compared to MI-treated hearts. SDF-1α shRNA, however, abolished VNS-improved LV function (Figure 10A–D, Figure S3).
[IMAGE OMITTED. SEE PDF]
Discussion
Our studies made three novel observations. First, we found that VNS-induced SDF-1α expression regulated angiogenesis in infarcted hearts. Second, ACh recovered SDF-1α/CXCR4 distribution along new branches during the formation of the vessel. Finally, SDF-1α expression was regulated by the ACh/m/nAChR/AKT/Sp1 signalling cascade.
Accumulated data have shown that vigorous inflammatory responses generally cause endothelial dysfunction in various human diseases, including MI. As a typical phenotype of endothelial dysfunction, the disruption of endothelial cell-branching morphogenesis and angiogenesis are evidently triggered by inflammatory cytokines such as IL-1β, IL-6, IFN-γ, and/or TNF-α.31,32 Indeed, IL-1β and IL-6 promote SDF-1α expression, while IFN-γ and TNF-α reduce its expression in various cells,31,32 indicating that inflammatory cytokines should positively and negatively control SDF-1α levels. Herein, accompanied by a decrease in these inflammatory cytokines, we further found that VNS induced SDF-1α expression in endothelial cells of infarcted hearts. This is in accordance with a recent report that SDF-1α acted as an anti-inflammatory chemokine during autoimmune inflammatory responses,33 suggesting that VNS and the vagal neurotransmitter ACh, which exert an anti-inflammatory effect, could have an independent role in SDF-1α expression. Initially, we provide novel evidence that ACh induces SDF-1α expression in CECs under noninflammatory conditions. More importantly, VNS-induced SDF-1α in infarcted hearts participated in the enhanced process of angiogenesis similar to SDF-1α-mediated microvascular network remodelling.34 Significantly, VNS induced the differential expression distribution characterized by SDF-1α in the epicardium and VEGF in the endocardium. SDF-1α has unique effects on promoting vascular reassembly in the infarcted heart,35 and the combined effect of epicardial SDF-1α and endocardial VEGF on cardiomyocyte replenishment initially occurs through the guidance of coronary sprouting.36 In summary, VNS could play a unique role in the angiogenesis of the infarcted heart by inducing SDF-1α and VEGF expression.
Indeed, ECs in the heart contain both m1 and m3-AChR apart from α7-nACh-R, the selective m1AChR inhibitor pirenzepine, and the selective m3AChR inhibitor 4-DAMP, which could better prove the role of VNS and ACh in SDF-1α expression and angiogenesis. Herein, we used the nonselective mAChR inhibitor atropine and found that atropine effectively eliminated VNS- and ACh-induced SDF-1α expression and angiogenesis, indirectly indicating that atropine could block m1 and m3AChRs. More accurate experiments in the future need to be designed to prove the relationship between ACh and m1/m3AChR in ECs. Meanwhile, using a selective α7-nACh-R inhibitor, memamylamine, we found that atropine and mecamylamine abolished the effect of VNS and ACh on SDF-1α expression and angiogenesis. In any case, we draw a preliminary conclusion that ACh induced SDF-1α expression and angiogenesis through m/n-AChR.
Published data have shown that NF-κB, PI3K/AKT, p38 MAPK, ERK1/2, and eNOS signalling can regulate SDF-1α expression in ECs, endothelial progenitor cells (EPCs), and mesenchymal stem cells.37–42 In fact, in infarcted hearts, apart from nutrient deficiency, hypoxia is an important characteristic that induces SDF-1α expression and cell migration through hypoxia inducible factor 1 alpha (HIF1α) and NF-κB p65.43–46 ACh acted as a cardioprotective agent by increasing HIF1a levels in the infarcted heart, suggesting that HIF1a-mediated SDF-1α expression may likely participate in the process of repairing the infarcted heart. Herein, under normoxic conditions, P13K/AKT signalling activated by ACh is involved in SDF-1α expression. Similar to a previous report that pAKT could phosphorylate transcription factor Sp1 and increase its activity,47 increased pAKT levels in ACh-treated HCAECs induced phosphorylation of Sp1, indicating that ACh promoted SDF-1α expression through the P13K/AKT-Sp1 signalling pathway.
In addition, increased LPS in the infarcted heart significantly inhibited angiogenesis and damaged the structure and function of the heart.24,25,48–50 After VNS, gene chip analysis showed a pronounced decrease in LPS-induced inflammatory responses in infarcted hearts in vivo, accompanied by an increase in angiogenesis.13 Herein, we found that ACh induced SDF-1α expression and tube formation in LPS-injured HCAECs.
CXCR4, as a receptor for SDF-1α, is the only chemokine receptor for which knockout mice die perinatally.51 Mice lacking CXCR4 show lethally defective cardiac ventricular septa and embryonic haematopoiesis and neurogenesis, a phenotype similar to that of SDF-1α knockout mice.52 Of interest, unlike previous results showing that LPS upregulated SDF-1α and CXCR4 expression in macrophages and fibroblast cells,53,54 LPS decreased SDF-1α and CXCR4 expression in HCAECs. ACh abolished the inhibitory effect of LPS on SDF-1α and CXCR4 expression, and a m/n-AChR blocker abolished these specific effects (Figures S4, S5 and S6). More importantly, ACh did not alter SDF-1α and CXCR4/7 distribution within HCAECs in proliferation medium (Figures S3, S4, and S5). However, under the vascular tube formation system, we found for the first time that ACh altered the distribution pattern of SDF-1α and its receptor CXCR4 in HCAECs. LPS reconstituted the distribution of SDF-1α around the nucleus accompanied by CXCR4, not CXCR7, leading to disorganization of endothelial cell-branching morphogenesis. ACh partially reversed these effects. Meanwhile, ACh-mediated SDF-1α redistribution and angiogenesis could be abolished by a m/n-AChR blocker. These results suggested that VNS and the vagal neurotransmitter ACh promoted angiogenesis and repaired the infarcted heart by altering the expression and distribution of SDF-1α/CXCR4.
Limitations
Clinically, VNS has attracted much attention due to its cholinergic anti-inflammatory and anti-adrenergic effects on MI and HF. Herein, we provide novel evidence that the effect of VNS on MI and HF is attributed to the unique synergy of epicardial SDF-1α and endocardial VEGF,13,14 especially in the coronary artery. In the past, we used the combined treatment of VEGF and SDF-1α genes to better repair MI,55 but how VNS cooperates with the expression pattern of VEGF and SDF-1α in the endocardium and epicardium to facilitate the repair of MI still needs to be further clarified in the future. In addition, using wired and radio-controlled pulse generators to stimulate the vagal nerve, the progression of MI and HF has been obviously delayed, with a better prognosis of HF.1–4 Thus, whether VNS in different ways is equivalent to driving synergetic expression patterns of epicardial SDF-1α and endocardial VEGF to facilitate the repair of MI still needs further clarification in the future. The solution of these problems will be more conducive to promoting the clinical research and application of VNS in the treatment of MI and HF.
Taken together, our studies demonstrated that VNS improves heart function by promoting angiogenesis in the infarcted heart via activation of SDF-1α signalling, which was regulated by the P13K/AKT-Sp1 signalling cascade (Figure 11).
[IMAGE OMITTED. SEE PDF]
Author contributions
Y.W., X.Y.L. and Y.L. carried out the main animal and cell experiments and drafted the manuscript. L.L.S. and Y.X.L. carried out the animal experiments. X.B. and L.Y. carried out protein detection; X.B. and Y.W. carried out the HCAEC cultures. L.Y.Y. and L.C. participated in the immunoassaying. J.X.Z. and S.J.C. carried out data evaluation. S.Y.C. and M.N.M. helped draft and revise the manuscript. J.M.T., J.L. and L.L.S. conceived of the study, participated in the experimental design and coordination of the study, and helped draft the manuscript. All authors read and approved the final manuscript.
Conflict of interest
The authors declare that they have no competing interests.
Funding
This study was supported by grants from the Hubei Provincial Technology Innovation Project (2018ACA162 and 2021DFE026 to J.M.T.), the Foundation of Hubei University of Medicine (HBMUPI201807 and FDFR201601 to J.M.T.), Hubei Province's Outstanding Medical Academic Leader Program, the National Natural Science Foundation of China (81670272 and 82270299 to J.M.T. and 82000285 to J.X.Z.) and the Natural Science Foundation of Hubei Province (2022CFB934 to L.L.S.). [Correction added on 28 September 2023, after first online publication: The funding number of ‘Natural Science Foundation of Hubei Province’ has been corrected from ‘2020CFB624 to Y.W’ to ‘2022CFB934 to L.L.S.’ in this version.]
Li P, Liu H, Sun P, Wang X, Wang C, Wang L, Wang T. Chronic vagus nerve stimulation attenuates vascular endothelial impairments and reduces the inflammatory profile via inhibition of the NF‐κB signaling pathway in ovariectomized rats. Exp Gerontol. 2016; 74: 43–55.
Li M, Zheng C, Sato T, Kawada T, Sugimachi M, Sunagawa K. Vagal nerve stimulation markedly improves long‐term survival after chronic heart failure in rats. Circulation. 2004; 109: 120–124.
Martelli D, Yao ST, McKinley MJ, McAllen RM. Reflex control of inflammation by sympathetic nerves, not the vagus. J Physiol. 2014; 592: 1677–1686.
Shen MJ, Shinohara T, Park HW, Frick K, Ice DS, Choi EK, Han S, Maruyama M, Sharma R, Shen C, Fishbein MC, Chen LS, Lopshire JC, Zipes DP, Lin SF, Chen PS. Continuous low level vagus nerve stimulation reduces stellate ganglion nerve activity and paroxysmal atrial tachyarrhythmias in ambulatory canines. Circulation. 2011; 123: 2204–2280.
Niccoli G, Scalone G, Lerman A, Crea F. Coronary microvascular obstruction in acute myocardial infarction. Eur Heart J. 2016; 37: 1024–1040.
Pillai S, Chellappan S. α7 nicotinic acetylcholine receptor subunit in angiogenesis and epithelial to mesenchymal transition. Curr Drug Targets. 2012; 13: 671–679.
Yu JG, Song SW, Shu H, Fan SJ, Liu AJ, Liu C, Guo W, Guo JM, Miao CY, Su DF. Baroreflex deficiency hampers angiogenesis after myocardial infarction via acetylcholine‐α7‐nicotinic ACh receptor in rats. Eur Heart J. 2013; 34: 2412–2420.
Pillay CV, Reid JV. Histochemical localization of acetylcholinesterase in the wall of cardiac blood vessels in the baboon, dog and vervet monkey. Basic Res Cardiol. 1982; 77: 213–219.
Akiyama T, Yamazaki T. Effects of right and left vagal stimulation on left ventricular acetylcholine levels in the cat. Acta Physiol Scand. 2001; 172: 11–16.
Coote JH. Myths and realities of the cardiac vagus. J Physiol. 2013; 591: 4073–4085.
Cooke JP, Ghebremariam YT. Endothelial nicotinic acetylcholine receptors and angiogenesis. Trends Cardiovasc Med. 2008; 18: 247–253.
Zhao M, He X, Bi XY, Yu XJ, Gil Wier W, Zang WJ. Vagal stimulation triggers peripheral vascular protection through the cholinergic anti‐inflammatory pathway in a rat model of myocardial ischemia/reperfusion. Basic Res Cardiol. 2013; 108: 345.
Luo B, Wu Y, Liu SL, Li XY, Zhu HR, Zhang L, Zheng F, Liu XY, Guo LY, Wang L, Song HX, Lv YX, Cheng ZS, Chen SY, Wang JN, Tang JM. Vagus nerve stimulation optimized the cardiomyocyte phenotype, sarcomere organization and energy metabolism in the infracted heart through FoxO3A‐VEGF signaling. Cell Death Dis. 2020; 11: 971.
Lv YX, Zhong S, Tang H, Luo B, Chen SJ, Chen L, Zheng F, Zhang L, Wang L, Li XY, Yan YW, Pan YM, Jiang M, Zhang YE, Wang L, Yang JY, Guo LY, Chen SY, Wang JN, Tang JM. VEGF‐A and VEGF‐B coordinate the arteriogenesis to repair the infarcted heart with vagus nerve stimulation. Cell Physiol Biochem. 2018; 48: 433–449.
Tang JM, Wang JN, Zhang L, Zheng F, Yang JY, Kong X, Guo LY, Chen L, Huang YZ, Wan Y, Chen SY. VEGF/SDF‐1 promotes cardiac stem cell mobilization and myocardial repair in the infracted heart. Cardiovasc Res. 2011; 91: 402–411.
Hu X, Dai S, Wu WJ, Tan W, Zhu X, Mu J, Guo Y, Bolli R, Rokosh G. Stromal cell derived factor‐1 alpha confers protection against myocardial ischemia/reperfusion injury: role of the cardiac stromal cell derived factor‐1 alpha CXCR4 axis. Circulation. 2007; 116: 654–663.
Saxena A, Fish JE, White MD, Yu S, Smyth JWP, Shaw RM, DiMaio JM, Srivastava D. Stromal cell‐derived factor‐1 alpha is cardioprotective after myocardial infarction. Circulation. 2008; 117: 2224–2231.
van Gils JM, Ramkhelawon B, Fernandes L, Stewart MC, Guo L, Seibert T, Menezes GB, Cara DC, Chow C, Kinane TB, Fisher EA. Endothelial expression of guidance cues in vessel wall homeostasis dysregulation under proatherosclerotic conditions. Arterioscler Thromb Vasc Biol. 2013; 33: 911–919.
Ceradini DJ, Kulkarni AR, Callaghan MJ, Tepper OM, Bastidas N, Kleinman ME, Capla JM, Galiano RD, Levine JP, Gurtner GC. Progenitor cell trafficking is regulated by hypoxic gradients through HIF‐1 induction of SDF‐1. Nat Med. 2004; 10: 858–864.
De Falco E, Porcelli D, Torella AR, Straino S, Iachininoto MG, Orlandi A, Truffa S, Biglioli P, Napolitano M, Capogrossi MC, Pesce M. SDF‐1 involvement in endothelial phenotype and ischemia‐induced recruitment of bone marrow progenitor cells. Blood. 2004; 104: 3472–3482.
Greenbaum A, Hsu YM, Day RB, Schuettpelz LG, Christopher MJ, Borgerding JN, Nagasawa T, Link DC. CXCL12 in early mesenchymal progenitors is required for hematopoietic stem cell maintenance. Nature. 2013; 495: 227–230.
MacArthur JW Jr, Purcell BP, Shudo Y, Cohen JE, Fairman A, Trubelja A, Patel J, Hsiao P, Yang E, Lloyd K, Hiesinger W, Atluri P, Burdick JA, Woo YJ. The sustained release of engineered stromal cell‐derived factor 1‐α from injectable hydrogels effectively recruits endothelial progenitor cells and preserves ventricular function after myocardial infarction. Circulation. 2013; 128: S79–S86.
Tang JM, Luo B, Xiao JH, Lv YX, Li XL, Zhao JH, Zheng F, Zhang L, Chen L, Yang JY, Guo LY, Wang L, Yan YW, Pan YM, Wang JN, Li DS, Wan Y, Chen SY. VEGF‐A promotes cardiac stem cell engraftment and myocardial repair in the infracted heart. Int J Cardiol. 2015; 183: 221–231.
Zhou H, Qian J, Li C, Li J, Zhang X, Ding Z, Gao X, Han Z, Cheng Y, Liu L. Attenuation of cardiac dysfunction by HSPA12B in endotoxin‐induced sepsis in mice through a PI3K‐dependent mechanism. Cardiovasc Res. 2011; 89: 109–118.
Jia Z, Huang Y, Ji X, Sun J, Fu G. Ticagrelor and clopidogrel suppress the NF‐κB signaling pathway to alleviate LPS‐induced dysfunction in vein endothelial cells. BMC Cardiovasc Disord. 2019; 19: 318.
DeCicco‐Skinner KL, Henry GH, Cataisson C, Tabib T, Gwilliam JC, Watson NJ, Bullwinkle EM, Falkenburg L, O'Neill RC, Morin A, Wiest JS. Endothelial cell tube formation assay for the in vitro study of angiogenesis. J Vis Exp. 2014; 91: [eLocator: e51312].
Griesinger CB, Richards CD, Ashmore JF. Apical endocytosis in outer hair cells of the mammalian cochlea. Eur J Neurosci. 2004; 20: 41–50.
Grauss RW, Winte EM, van Tuyn J, Pijnappels DA, Steijn RV, Hogers B, van der Geest RJ, de Vries AA, Steendijk P, van der Laarse A, Gittenberger‐de Groot AC, Schalij MJ, Atsma DE. MSCs from ischemic heart disease patients improve left ventricular function after AMI. Am J Physiol ‐ Heart Circ Physiol. 2007; 293: H2438–H2447.
Tang JM, Shi N, Dong K, Brown SA, Coleman AE, Boegehold MA, Chen SY. Response gene to complement 32 maintains blood pressure homeostasis by regulating α‐adrenergic receptor expression. Circ Res. 2018; 123: 1080–1090.
Bromage DI, Davidson SM, Yellon DM. Stromal derived factor 1α: a chemokine that delivers a two‐pronged defense of the myocardium. Pharmacol Ther. 2014; 143: 305–315.
Blaine TA, Cote MA, Proto A, Mulcahey M, Lee FY, Bigliani LU. Interleukin‐1β stimulates stromal‐derived factor‐1α expression in human subacromial bursa. J Orthop Res. 2011; 29: 1695–1699.
Salvucci O, Basik M, Yao L, Bianchi R, Tosato G. Evidence for the involvement of SDF‐1 and CXCR4 in the disruption of endothelial cell‐branching morphogenesis and angiogenesis by TNF‐alpha and IFN‐gamma. J Leukoc Biol. 2004; 76: 217–226.
Mousavi A. CXCL12/CXCR4 signal transduction in diseases and its molecular approaches in targeted‐therapy. Immunol Lett. 2020; 217: 91–115.
Krieger JR, Ogle ME, McFaline‐Figueroa J, Segar CE, Temenoff JS, Botchwey EA. Spatially localized recruitment of anti‐inflammatory monocytes by SDF‐1α‐releasing hydrogels enhances microvascular network remodelling. Biomaterials. 2016; 77: 280–290.
Das S, Goldstone AB, Wang H, Farry J, D'Amato G, Paulsen MJ, Eskandari A, Hironaka CE, Phansalkar R, Sharma B, Rhee S, Shamskhou EA, Agalliu D, de Jesus Perez V, Woo YJ, Red‐Horse K. A unique collateral artery development program promotes neonatal heart regeneration. Cell. 2019; 176: 1128–1142.
Marín‐Juez R, El‐Sammak H, Helker CSM, Kamezaki A, Mullapuli ST, Bibli SI, Foglia MJ, Fleming I, Poss KD, Stainier DYR. Coronary revascularization during heart regeneration is regulated by epicardial and endocardial cues and forms a scaffold for cardiomyocyte repopulation. Dev Cell. 2019; 51: 503–515.e4.
Hsieh YY, Shen CH, Huang WS, Chin CC, Kuo YH, Hsieh MC, Yu HR, Chang TS, Lin TH, Chiu YW, Chen CN, Kuo HC, Tung SY. Resistin‐induced stromal cell‐derived factor‐1 expression through toll‐like receptor 4 and activation of p38 MAPK/NFκB signaling pathway in gastric cancer cells. J Biomed Sci. 2014; 21: 59.
Kazakov A, Hall R, Jagoda P, Bachelier K, Mueller‐Best P, Semenov A, Lammert F, Böhm M, Laufs U. Inhibition of endothelial nitric oxide synthase induces and enhances myocardial fibrosis. Cardiovasc Res. 2013; 100: 211–221.
McCorkell KA, May MJ. Noncanonical NF‐κB activation and SDF‐1 expression in human endothelial cells. Methods Mol Biol. 2015; 1280: 155–180.
Picinich SC, Glod JW, Banerjee D. Protein kinase C zeta regulates interleukin‐8‐mediated stromal‐derived factor‐1 expression and migration of human mesenchymal stromal cells. Exp Cell Res. 2010; 316: 593–602.
Poh KK, Lee PSS, Djohan AH, Galupo MJ, Songco GG, Yeo TC, Tan HC, Richards AM, Ye L. Transplantation of endothelial progenitor cells in obese diabetic rats following myocardial infarction: role of thymosin beta‐4. Cell. 2020; 9: 949.
Stępniewski J, Tomczyk M, Andrysiak K, Kraszewska I, Martyniak A, Langrzyk A, Kulik K, Wiśniewska E, Jeż M, Florczyk‐Soluch U, Polak K, Podkalicka P, Kachamakova‐Trojanowska N, Józkowicz A, Jaźwa‐Kusior A, Dulak J. Human induced pluripotent stem cell‐derived cardiomyocytes, in contrast to adipose tissue‐derived stromal cells, efficiently improve heart function in murine model of myocardial infarction. Biomedicine. 2020; 8: 578.
Loh SA, Chang EI, Galvez MG, Thangarajah H, El‐Ftesi S, Vial IN, Lin DA, Gurtner GC. SDF‐1 alpha expression during wound healing in aged individuals is HIF dependent. Plast Reconstr Surg. 2009; 123: 65 s–75s.
Xue Y, Li Z, Wang Y, Zhu X, Hu R, Xu W. Role of the HIF‐1α/SDF‐1/CXCR4 signaling axis in accelerated fracture healing after craniocerebral injury. Mol Med Rep. 2020; 22: 2767–2774.
Kew RR, Penzo M, Habiel DM, Marcu KB. The IKKα‐dependent NF‐κB p52/RelB noncanonical pathway is essential to sustain a CXCL12 autocrine loop in cells migrating in response to HMGB1. J Immunol. 2012; 188: 2380–2386.
Yin W, Ma L, Zhang J, Huang K, Yang Q, Guo YY, Liu SB, Liu YH, Wu YM. The migration of neural progenitor cells mediated by SDF‐1 is NF‐κB/HIF‐1α‐dependent upon hypoxia. CNS Neurosci Ther. 2013; 19: 145–153.
Lin HH, Lai SC, Chau LY. Heme oxygenase‐1/carbon monoxide induces vascular endothelial growth factor expression via p38 kinase‐dependent activation of Sp1. J Biol Chem. 2011; 286: 3829–3838.
Adamo L, Staloch LJ, Rocha‐Resende C, Matkovich SJ, Jiang W, Bajpai G, Weinheimer CJ, Kovacs A, Schilling JD, Barger PM, Bhattacharya D, Mann DL. Modulation of subsets of cardiac B lymphocytes improves cardiac function after acute injury. JCI Insight. 2018; 3: [eLocator: e120137].
Chao W. Toll‐like receptor signaling: a critical modulator of cell survival and ischemic injury in the heart. Am J Physiol ‐ Heart Circ Physiol. 2009; 296: H1–H12.
Lin ZH, Li YC, Wu SJ, Zheng C, Lin YZ, Lian H, Lin WQ, Lin JF. Eliciting α7‐nAChR exerts cardioprotective effects on ischemic cardiomyopathy via activation of AMPK signaling. J Cell Mol Med. 2019; 23: 4746–4758.
Lapidot T. Mechanism of human stem cell migration and repopulation of NOD/SCID and B2mnull NOD/SCID mice. The role of SDF‐1/CXCR4 interactions. Ann N Y Acad Sci. 2001; 938: 83–95.
Xu J, Mora A, Shim H, Stecenko A, Brigham KL, Rojas M. Role of the SDF‐1/CXCR4 axis in the pathogenesis of lung injury and fibrosis. Am J Respir Cell Mol Biol. 2007; 37: 291–299.
Ding J, Hori K, Zhang R, Marcoux Y, Honardoust D, Shankowsky HA, Tredget EE. Stromal cell‐derived factor 1 (SDF‐1) and its receptor CXCR4 in the formation of postburn hypertrophic scars (HTSs). Wound Repair Regen. 2011; 19: 568–578.
Yu L, Yu L, Quynhchi P, Wang TTY. Transcriptional and translational uncoupling in the regulation of CXCL12 and its receptors CXCR4 and 7 in THP‐1 monocytes and macrophages. Immun Inflamm Dis. 2018; 6: 106–116.
Tang JM, Wang JN, Zheng F, Kong X, Guo L, Yang JY, Zhang L, Huang YZ. Combination of chemokine and angiogenic factor genes and mesenchymal stem cells could enhance angiogenesis and improve cardiac function after acute myocardial infarction in rats. Mol Cell Biochem. 2010; 339: 107–118.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
© 2023. This work is published under http://creativecommons.org/licenses/by-nc/4.0/ (the "License"). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
Aims
We aim to explore the role and mechanism of vagus nerve stimulation (VNS) in coronary endothelial cells and angiogenesis in infarcted hearts.
Methods and results
Seven days after rat myocardial infarction (MI) was prepared by ligation of the left anterior descending coronary artery, the left cervical vagus nerve was treated with electrical stimulation 1 h after intraperitoneal administration of the α7‐nicotinic acetylcholine inhibitor mecamylamine or the mAChR inhibitor atropine or 3 days after local injection of Ad‐shSDF‐1α into the infarcted heart. Cardiac tissue acetylcholine (ACh) and serum ACh, tumour necrosis factor α (TNF‐α), interleukin 1β (IL‐1β) and interleukin 6 (IL‐6) levels were detected by ELISA to determine whether VNS was successful. An inflammatory injury model in human coronary artery endothelial cells (HCAECs) was established by lipopolysaccharide and identified by evaluating TNF‐α, IL‐1β and IL‐6 levels and tube formation. Immunohistochemistry staining was performed to evaluate CD31‐positive vessel density and stromal cell‐derived factor‐l alpha (SDF‐1α) expression in the MI heart in vivo and the expression and distribution of SDF‐1α, C‐X‐C motif chemokine receptor 4 and CXCR7 in HCAECs in vitro. Western blotting was used to detect the levels of SDF‐1α, V‐akt murine thymoma viral oncogene homolog (AKT), phosphorylated AKT (pAKT), specificity protein 1 (Sp1) and phosphorylation of Sp1 in HCAECs. Left ventricular performance, including left ventricular systolic pressure, left ventricular end‐diastolic pressure and rate of the rise and fall of ventricular pressure, should be evaluated 28 days after VNS treatment. VNS was successfully established for MI therapy with decreases in serum TNF‐α, IL‐1β and IL‐6 levels and increases in cardiac tissue and serum ACh levels, leading to increased SDF‐1α expression in coronary endothelial cells of MI hearts, triggering angiogenesis of MI hearts with increased CD31‐positive vessel density, which was abolished by the m/nAChR inhibitors mecamylamine and atropine or knockdown of SDF‐1α by shRNA. ACh promoted SDF‐1α expression and its distribution along with the branch of the formed tube in HCAECs, resulting in an increase in the number of tubes formed in HCAECs. ACh increased the levels of pAKT and phosphorylation of Sp1 in HCAECs, resulting in inducing SDF‐1α expression, and the specific effects could be abolished by mecamylamine, atropine, the PI3K/AKT blocker wortmannin or the Sp1 blocker mithramycin. Functionally, VNS improved left ventricular performance, which could be abolished by Ad‐shSDF‐1α.
Conclusions
VNS promoted angiogenesis to repair the infarcted heart by inducing SDF‐1α expression and redistribution along new branches during angiogenesis, which was associated with the m/nAChR‐AKT‐Sp1 signalling pathway.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Details
1 Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, PR China, Department of Pathology, Renmin Hospital, Hubei University of Medicine, Shiyan, PR China
2 Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, PR China
3 Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, PR China, Department of Anesthesiology, Institute of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, PR China
4 Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, PR China, Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan, PR China
5 Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, PR China, Experimental Medical Center, Guoyao‐Dong Feng Hospital, Hubei University of Medicine, Shiyan, PR China
6 Experimental Medical Center, Guoyao‐Dong Feng Hospital, Hubei University of Medicine, Shiyan, PR China
7 Department of Surgery, University of Missouri, Columbia, Missouri, USA
8 Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, PR China, Institute of Basic Medical Sciences, Institute of Biomedicine, Hubei University of Medicine, Shiyan, PR China
9 Department of Anesthesiology, Institute of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, PR China