Open Access

Intermedin1‑53 enhances angiogenesis and attenuates adverse remodeling following myocardial infarction by activating AMP‑activated protein kinase

  • Authors:
    • Kankai Chen
    • Meiling Yan
    • Yongguang Li
    • Zhifeng Dong
    • Dong Huang
    • Jingbo Li
    • Meng Wei
  • View Affiliations

  • Published online on: February 10, 2017     https://doi.org/10.3892/mmr.2017.6193
  • Pages: 1497-1506
  • Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Adverse ventricular remodeling is a maladaptive response to acute loss of myocardium and an important risk factor for heart failure following myocardial infarction (MI). Intermedin (IMD) is a novel member of the calcitonin/calcitonin gene‑related peptide family, which may possess potent cardioprotective properties. The aim of the present study was to determine whether IMD1‑53, a mature bioactive form of IMD, may promote therapeutic angiogenesis within the infarcted myocardium, therefore attenuating adverse ventricular remodeling post‑MI. The present study observed that treatment with IMD1‑53 promoted proliferation, migration and tube formation of primary cultured myocardial microvascular endothelial cells (MMVECs). In a rat model of MI, chronic administration of IMD1‑53 increased capillary density in the peri‑infarct zone, attenuated ventricular remodeling and improved cardiac performance post‑MI. Treatment with IMD1‑53 also significantly increased the expression levels of phosphorylated‑AMP‑activated protein kinase (AMPK) and the subsequent activation of endothelial nitric oxide synthase in MMVECs and post‑MI rat myocardium, without a significant influence on the expression of vascular endothelial growth factor. Notably, the in vitro effects of IMD1‑53 on angiogenesis and the in vivo effects of IMD1‑53 on post‑MI ventricular remodeling were largely abrogated by the co‑administration of compound C, an AMPK inhibitor. In conclusion, the present study demonstrated that IMD1‑53 could attenuate adverse ventricular remodeling post‑MI via the promotion of therapeutic angiogenesis, possibly through the activation of AMPK signaling.

Introduction

Ventricular remodeling, which is characterized by progressive dilation and deterioration in cardiac performance, is a critical process underlying the progression to heart failure and subsequent mortality following myocardial infarction (MI). As a result, the attenuation of adverse myocardial remodeling is one of the most important aspects for improving prognosis following MI (1). However, despite previous advances in reperfusion therapy and optimized pharmacological treatments, left ventricular (LV) remodeling is still observed in a substantial proportion of patients, and progression to heart failure can still occur in up to one-third of patients who have had a MI (2,3). These data suggest that the current clinical treatments targeting myocardial remodeling post-MI remain inadequate. The promotion of therapeutic angiogenesis has been demonstrated to be a promising approach to ameliorate adverse myocardial remodeling (4).

Intermedin (IMD), also known as adrenomedullin 2, is a novel member of the calcitonin/calcitonin gene-related peptide (CGRP) family. The human IMD gene encodes a prepropeptide of 148 amino acids (preproIMD); proteolytic cleavage of preproIMD yields a series of biologically active C-terminal fragments. Previous studies have revealed that an endogenous degraded fragment termed IMD1–53, which contains cleavage sites located between two basic amino acids at Arg93-Arg94, possessed potent cardioprotective activities in multiple physiopathological processes, including ischemia/reperfusion injury (5,6), cardiac fibrosis (7) and cardiac hypertrophy (8,9). However, the mechanisms underlying the cardioprotective effects of IMD require further elucidation. Notably, IMD was also reported to serve a role in the regulation of angiogenesis in vitro and in vivo (10,11). However, whether IMD1–53 may promote therapeutic angiogenesis within the infarcted myocardium, and therefore attenuate adverse myocardial remodeling post-MI, has not been investigated. To test this hypothesis, the present study investigated the effects of recombinant IMD1–53 on angiogenesis in primary cultured myocardial microvascular endothelial cells (MMVECs) and in a rat model of MI. The results suggested that IMD1–53 attenuated adverse ventricular remodeling post-MI by promoting therapeutic angiogenesis, possibly through the activation of AMP-activated protein kinase (AMPK) signaling.

Materials and methods

Materials

IMD1–53 peptide was purchased from Phoenix Pharmaceuticals, Inc. (Burlingame, CA, USA). Dulbecco's modified Eagle's medium (DMEM) and fetal bovine serum (FBS) were purchased from Gibco (Thermo Fisher Scientific, Inc., Waltham, MA USA). Phosphorylated (p)-AMPKαThr-172 (cat. no. 2535), AMPKα (cat. no. 2532), p-AktSer-473 (cat. no. 4060), and p-endothelial nitric oxide synthase (p-eNOS)Ser-1179 (cat. no. 9570) antibodies were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA). Anti-Akt (cat. no. sc-8312), anti-eNOS (cat. no. sc-654), anti-GAPDH (cat. no. sc-25778) and anti-VEGF (cat. no. sc-152), anti-CD31 (cat. no. sc-1505) anti-CD34 (cat. no. sc-7045) antibodies were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). Compound C (Comp C) was purchased from Toronto Research Chemicals Inc. (North York, ON, Canada). Goat anti-rabbit (cat. no. sc-2030) and goat anti-mouse (cat. no. sc-3791) secondary antibodies were purchased from Santa Cruz Biotechnology, Inc.

For all experiments concerning IMD1–53, the same volume of PBS was used as a vehicle. For experiments concerning Comp C, the same volume of DMSO was used as a vehicle.

Isolation and identification of MMVECs

Male Wistar rats (n=16; 4–6 weeks; 80–100 g) were used for the isolation of primary MMVECs. The rats were housed at room temperature with a 12/12 h light/dark cycle and free access to food and water. The animal protocol was approved by the Animal Care Committee of Shanghai Jiao Tong University (Shanghai, China). Rats were sacrificed with an overdose of sodium pentobarbital (180 mg/kg) and heparinized by intraperitoneal injection of sodium heparin (500 units/0.1 kg). Following thoracotomy, the heart was rapidly dislodged and washed in PBS. The atria, right ventricle, epicardial tissue and visible connective tissue were carefully removed, and the remaining myocardial tissue was washed in PBS prior to cutting into 1 mm3 sections without visible vessels. Myocardial tissues were seeded onto culture plates that were pre-coated with rat tail tendon gelatin and subsequently incubated at 37°C in a humidified atmosphere containing 5% CO2 for 30 min. Tissues were cultured in DMEM containing 4,500 mg/l D-glucose and supplemented with 20% FBS, 50 U/ml heparin, 100 U/ml penicillin and 100 µg/ml streptomycin. Tissue sections were discarded after the cells began to grow, and the medium was replaced at 72 h intervals. MMVECs were identified by typical ‘cobblestone’ appearance and positive CD31 and CD34 immunostaining. MMVECs at the second passage were used for experiments. The cells were grown to 80–90% confluence and were used in subsequent experimental analyses.

Immunostaining

MMVECs were plated on a 12×12 mm plate, fixed by methanol for 15 min at −20°C, blocked with 10% normal goat serum (cat. no. 31872; Gibco; Thermo Fisher Scientific, Inc.) for 30 min at room temperature, incubated with CD31 (cat. no. sc-1505; Santa Cruz Biotechnology, Inc.) and CD34 (cat. no. sc-7045; Santa Cruz Biotechnology, Inc.) antibodies at 1:200 overnight at 4°C, and then incubated with goat anti-mouse secondary antibody (cat. no. sc-3791; Santa Cruz Biotechnology, Inc.) at 1:1,500 for 1 h at room temperature. Finally, DAB working solution diluted by H2O2 was added and the MMVECs were observed under a light microscope (Olympus IX71; Olympus Corporation, Tokyo, Japan).

Cell proliferation assay

The proliferation rate of MMVECs was assessed using the MTT assay (Sigma-Aldrich; Merck Millipore, Darmstadt, Germany). The cells were seeded into 96-well plates at a density of 2×103 cells/well and incubated in DMEM containing 10% FBS at 37°C with 5% CO2 for 24 h. IMD1–53, Comp C or IMD1–53+Comp C was added to the medium at different concentrations (0, 10, 20, 40, 80, 160 nm of IMD and/or 20 µmol compound C) at 0 h. Following 0, 8, 16, 24 and 48 h incubation, 10 µl of MTT was added to each well and the cells were incubated at 37°C for 4 h. Following incubation, the supernatant fluid was removed and 100 µl dimethyl sulfoxide was added to each well. The cells were incubated for 10 min and the absorbance at 490 nm was measured with an Epoch Microplate Spectrophotometer (BioTek Instruments, Inc., Winooski, VT, USA).

Migration assay

MMVEC migration rate was assessed using the Boyden Chemotaxis Chamber assay (Neuro Probe, Inc., Gaithersburg, MD, USA). The cells were trypsinized and resuspended in 10% DMEM containing 10% FBS. IMD1–53 (80 nmol) and/or Comp C (20 µmol) were added to the wells in the lower chamber, and 1.5×104 cells (200 µl/well) were added into the upper chamber. Cells migrating through the filter were fixed in 4% paraformaldehyde for 10 min and subsequently stained with 0.1% crystal violet stain solution (Merck Millipore) for 30 min. Five random microscopic fields per well were quantified using an Olympus IX71 fluorescence microscope (Olympus Corporation). Each experiment was repeated three times.

Tube formation assay

Tube formation of MMVECs was assessed using a Matrigel assay (BD Biosciences, Franklin Lakes, NJ, USA). Matrigel was chilled at 4°C overnight, melted prior to use and then quickly added (70 µl/well) to 96-well plates using a pre-chilled pipette. The plates were incubated at 37°C in 5% CO2 for 1 h. The cells were seeded into the plates at a density of 1×104 cells/well and incubated for 12 h in the presence or absence of IMD1–53 (80 nmol) and/or Comp C (20 µmol) at 37°C. Tube formation was observed and images were captured using an Olympus 1X71 microscope (Olympus Corporation). Images were processed using Image-Pro Plus software 6.0 (Media Cybernetics, Inc., Rockville, MD, USA) to calculate the degree of tube formation by measuring the length of tubes from five randomly selected fields (magnification, ×200) from each well. Each experiment was repeated three times.

Western blot analysis

Cell or tissue samples were homogenized as follows: Freshly frozen myocardial tissue samples were ground into small pieces (~1×1×1 mm) in liquid nitrogen with a mortar and a pestle. The samples were transferred to microcentrifuges containing radioimmunoprecipitation lysis buffer (~150 µl per 10 mg tissue; Beyotime Institute of Biotechnology, Haimen, China) and 1 mM phenylmethylsulfonyl fluoride. The samples were then kept on ice for 1 h, vortexing every 10 min. The lysates were then centrifuged at 10,000 × g for 30 min at 4°C, and supernatants were removed for immediate western blot analysis. Protein samples (5 mg/ml) were loaded onto a 10% Bis-Tris gel; following electrophoresis, separated proteins were transferred to a polyvinylidene fluoride membrane for 1 h. The membranes were then incubated with blocking solution (5% skim milk) for 1–2 h at room temperature, followed by three washes with TBS-Tween-20 (TBST; 0.1% Tween; 10 min each). Membranes were incubated with one of the following primary antibodies: Anti-AMPK (1:800); anti-p-AMPKThr172 (1:1,000); anti-Akt (1:1,500); anti-p-Aktser473 (1:1,000); anti-eNOS (1:500); anti-p-eNOSser1117 (1:500); anti-vascular endothelial growth factor (VEGF; 1:1,000); and anti-GAPDH (1:8,000) at 4°C overnight. Primary antibodies were recovered and the membranes were washed three times with TBST (10 min each), followed by incubation with horseradish peroxidase-conjugated goat anti-rabbit immunoglobulin G (IgG; cat. no. sc-2030; 1:2,000) secondary antibody at room temperature for 1 h. Subsequently, the membranes were washed five times with TBST (5 min/wash), and the bands were detected using Western Blotting Luminol Reagent (Santa Cruz Biotechnology, Inc.) and quantified by densitometric analysis of digitized autoradiograms using Quantity One (version 4.6.2) software (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Each immunoblotting experiment was repeated three times, and the averages of the results were calculated.

Animal model

A total of 70 male Sprague-Dawley rats (age, 8 weeks; weight, 250–280 g; Experimental Animal Center, Fudan University, Shanghai, China) were used in the present study, and were maintained at room temperature with a 12/12 h light/dark cycle and free access to food and water. The animal research study protocol was in compliance with the Guide for the Care and Use of Laboratory Animals published by the National Institutes of Health (NIH Publication no. 85–23, revised 1996) and approved by the Animal Care Committee of Shanghai Sixth Hospital, Shanghai Jiao Tong University. All rats were acclimated for 2 weeks prior to experimentation. All rats were anesthetized by intraperitoneal injection of pentobarbital sodium (60 mg/kg) and ventilated using a DW-200 animal respirator (Shanghai Alcott Biotech Co., Ltd., Shanghai, China) with room air following endotracheal intubation. Electrocardiogram (ECG) lead II was continuously monitored during the experiment. Heart exposure was performed by a thoracotomy at the left fourth intercostal space. The left anterior descending (LAD) coronary artery was ligated using a size 6–0 Prolene polypropylene suture, 1–2 mm below the tip of the left atrial appendage. LAD ligation was confirmed by a color change of the myocardium and an elevation of the ST segment on the ECG. A total of 4 weeks after LAD ligation, all the surviving rats were sacrificed by injection of 180 mg/kg pentobarbital sodium overdose.

Animal protocols

A total of 70 Sprague-Dawley rats were randomly divided and assigned to five treatment groups: i) Sham group (n=10), which underwent thoracotomy and incision of the pericardial sac without LAD ligation; ii) MI group (n=15), which underwent LAD ligation without any treatment; iii) IMD group (n=15), which contained MI rats that received subsequent intraperitoneal injections of IMD1–53 (10 nmol/kg/day) starting 3 days after LAD ligation; iv) IMD+Comp C group (n=15), which contained MI rats that received subsequent intraperitoneal injections of IMD1–53 (10 nmol/kg/day) and Comp C (5 mg/kg/day) starting 3 days after LAD ligation; and v) Comp C group (n=15), which contained MI rats that received subsequent intraperitoneal injections of Comp C (5 mg/kg) starting 3 days after LAD ligation.

Echocardiography

An echocardiographic examination was performed on day 28 following the induction of MI using a standard Acuson Sequoia 512 ultrasound system equipped with a 15 MHz probe (Siemens AG, Munich, Germany). Two-dimensional and motion-mode echocardiography were performed. The LV end-diastolic diameter (LVEDD) and the LV end-systolic diameter (LVESD) were measured. In addition, the LV ejection fraction (LVEF) and fractional shortening (FS) were calculated as previously described (12). All measurements from five consecutive cardiac cycles were averaged and analyzed by a single observer that was blinded to the treatment protocol.

Histological analysis

Under deep anesthesia with pentobarbital sodium (intraperitoneal injection at 100 mg/kg), the heart was excised and cut into 2-mm-thick transverse slices, which were then fixed in 10% formalin solution at room temperature for 24 h. The samples were subsequently embedded in paraffin and sectioned (4 µm). Immunohistochemical analysis was performed as previously described (13). Tissue sections were then deparaffinized using xylene and graded ethanol and were blocked in 10% goat serum (Gibco, Thermo Fisher Scientific, Inc.) for 30 min at 37°C. Samples were incubated at 4°C overnight with primary rabbit polyclonal antibodies against CD31 (ab28364) and VEGF (ab46154; Abcam, Cambridge). Sections were subsequently treated with secondary goat polyclonal secondary antibody against rabbit IgG (ab97047; Abcam). All sections were counterstained with hematoxylin. Immunoreactivity for CD31 and VEGF was measured using a fluorescence microscope and the Image-Pro Plus 4.0 analysis system (Media Cybernetics, Inc.). Capillaries were identified by positive staining for CD31. Results were expressed as the average amount of capillaries per ×10 field. Masson's trichrome stain was performed in the border area to evaluate collagen deposition.

Statistical analysis

Statistical analysis of the data was conducted using the statistical software SPSS version 13.0 (SPSS, Inc., Chicago, IL, USA). Experimental data were expressed as the mean ± standard deviation and analyzed using one-way ANOVA followed by a least-significant difference corrected multiple comparison test. P<0.05 was considered to indicate a statistically significant difference.

Results

IMD1–53 increases proliferation of MMVECs in a dose-dependent manner

The effects of IMD1–53 on the proliferation of MMVECs were examined using the MTT assay. The results indicated that IMD1–53 was able to promote the proliferation of MMVECs in a dose-dependent manner with the maximum effect observed at 80 nmol (P<0.05 vs. 0 nmol control, Fig. 1A). In addition, co-administration of Comp C, a well-known inhibitor of AMPK, significantly attenuated the effects of IMD1–53 on the proliferation of MMVECs (P<0.05 vs. IMD at 8, 16, 24 and 48 h; Fig. 1B), whereas treatment with Comp C alone had no significant influence on the proliferation of MMVECs (P>0.05, Fig. 1B).

IMD1–53 promotes migration and tube formation in MMVECs

The effects of IMD1–53 on endothelial cell migration were further evaluated by a modified Boyden chamber assay (Fig. 2). The results indicated that the optimal dose (80 nmol, as shown in the MTT assay) of IMD1–53 treatment stimulated the migration of MMVECs (P<0.05 vs. control, Fig. 2A and C). Co-treatment with Comp C (20 µmol) partially suppressed the pro-migratory effect of IMD1–53 (P<0.05 vs. IMD, Fig. 2A and C).

To examine the effects of IMD1–53 on the differentiation of cardiovascular endothelial cells into vascular structures, MMVECs were plated at 1×104 cells/well in the presence or absence of IMD1–53 (80 nmol) and/or Comp C (20 µmol) and tube formation was detected microscopically. A total of 24 h after plating, the MMVECs treated with IMD1–53 formed a significantly more extensive tube network compared with the control group (P<0.05). The effects of IMD1–53 on tube formation were reduced in MMVECs that were also pretreated with Comp C (P<0.05 vs. IMD group). Treatment with Comp C alone significantly decreased tube formation as compared with the control group (P<0.05, Fig. 2B and D).

IMD1–53 stimulates activation of the AMPK-Akt-eNOS signaling pathway in MMVECs

Since AMPK is involved in the regulation of angiogenesis (14) and IMD1–53 was recently reported to increase AMPK activation in rat cardiomyocytes (9), the present study aimed to investigate whether AMPK is involved in IMD1–53-induced angiogenesis in MMVECs. MMVECs were exposed to IMD1–53 (80 nmol) for 2 h; subsequently, the total and phosphorylated protein levels of AMPKα, Akt and eNOS were analyzed by western blot analysis. As shown in Fig. 3, the expression levels of p-AMPKα, p-Akt and p-eNOS were all significantly increased by IMD1–53 treatment (P<0.05 vs. control, Fig. 3). Co-treatment with the Comp C suppressed the ability of IMD1–53 to activate either Akt or eNOS, as measured by the relative expression levels of p-Akt and p-eNOS (P<0.05 vs. IMD, Fig. 3), whereas treatment with Comp C alone slightly but significantly decreased the phosphorylated levels of Akt and eNOS (P<0.05, respectively; Fig. 3) and significantly decreased the phosphorylated levels of AMPKα (P<0.05; Fig. 3).

IMD1–53 increases capillary density in post-MI rat heart

To assess the effects of IMB of angiogenesis in vivo, myocardium capillary density tests were performed. Compared with the sham group, induction of MI significantly increased the capillary density in the peri-infarct zone (P<0.05, Fig. 4A and C). Treatment with IMD1–53 significantly increased capillary density in the peri-infarct zone of post-MI rat myocardium (P<0.05 vs. MI, Fig. 4A and C). Co-administration of Comp C mitigated the effects of IMD1–53 on capillary density in the peri-infarct zone (P<0.05 vs. IMD, Fig. 4A and C). Compared with the MI group, administration of Comp C alone slightly decreased capillary density in the peri-infarct zone (P<0.05, Fig. 4A and C). Compared to the sham group, the expression levels of VEGF were increased following induction of MI (P<0.05, Fig. 4B and D). However, the administration of IMD1–53, Comp C or a combination of the two had no significant effect on VEGF expression in the peri-infarct zone post-MI (P>0.05 vs. MI group, Fig. 4B and D).

IMD1–53 attenuates ventricular remodeling post-MI

To evaluate the effects of IMD1–53 on ventricular remodeling post-MI, Masson's trichrome staining was performed on all groups 4 weeks post-MI induction (Fig. 5). The results demonstrated that treatment with IMD1–53 markedly suppressed elongation of the infarct wall segment and dilation of the LV, as demonstrated by cross-section morphology (Fig. 5A) and calculated by infarct size post-MI (P<0.05 vs. MI group; Fig. 5B). The effects of IMD1–53 on ventricular remodeling were abrogated by co-administration of Comp C (P<0.05 vs. IMD, Fig. 5), whereas Comp C alone had no significant effect on myocardial infarct size post-MI (P>0.05 vs. MI, Fig. 5).

IMD1–53 improves cardiac function post-MI

A total of 4 weeks after MI induction, cardiac geometry and performance were evaluated using echocardiography (Table I). Compared with the sham group, significantly decreased LVEF and FS, along with increased LVESD and LVEDD, were observed in the MI group (P<0.05, Table I). In IMD-treated MI rats, LVEF and FS were significantly improved, whereas LVESD and LVEDD were significantly reduced, compared with the MI group (P<0.05, Table I). Co-administration of Comp C abrogated the effects of IMD1–53 on LVEF, FS, LVESD and LVEDD (P<0.05 vs. IMD, Table I), whereas Comp C alone had no significant effects on cardiac geometry and performance (P>0.05, Table I). Neither IMD1–53 nor Comp C administration had significant effects on survival rate of rats post-MI (P>0.05 vs. MI group; Table I).

Table I.

Representative motion-mode echocardiograms obtained 4 weeks post-MI induction.

Table I.

Representative motion-mode echocardiograms obtained 4 weeks post-MI induction.

ParameterShamMIIMDIMD+Comp CComp C
Number1015151515
Survival1011121110
LVESD (mm) 3.47±0.28a6.11±0.31 4.52±0.27a 5.84±0.33b6.24±0.28
LVEDD (mm) 5.83±0.36a8.28±0.44 6.42±0.42a 7.61±0.49b8.48±0.47
LVEF (%) 81.3±5.38a46.8±5.14 65.2±6.03a 53.7±5.93b44.9±5.26
FS (%) 75.6±4.97a44.9±3.71 66.5±4.89a 52.2±3.87b43.7±3.54

{ label (or @symbol) needed for fn[@id='tfn1-mmr-15-04-1497'] } Summary data of echocardiographic measurements in each group of animals. Data are expressed as the mean ± standard deviation.

a P<0.05 vs. MI group

b P<0.05 vs. IMD-treated group. Comp C, compound C; FS, fractional shortening; IMD, intermedin; LVEDD, left ventricular end-diastolic diameter; LVEF, left ventricular ejection fraction; LVESD, left ventricular end-systolic diameter; MI, myocardial infarction.

IMD1–53 stimulates activation of the AMPK-Akt-eNOS signaling pathway in post-MI rat myocardium

Western blot analysis was used to evaluate the activation of AMPK, Akt and eNOS, and the expression levels of VEGF in rat myocardium (Fig. 6). The results indicated that chronic IMD1–53 treatment resulted in significantly increased activation (as measured by phosphorylation) of AMPK, Akt and eNOS in post-MI rat myocardium (P<0.05 vs. MI, Fig. 6B-D), whereas it had no significant effect on the expression of VEGF (P>0.05 vs. MI, Fig. 6A). Co-administration of Comp C abrogated the effects of IMD1–53 on AMPK, Akt and eNOS phosphorylation (P<0.05 vs. IMD, Fig. 6B-D), whereas Comp C alone slightly decreased levels of p-AMPK, p-Akt and p-eNOS (P<0.05 vs. MI, Fig. 6B-D).

Discussion

Using in vitro and in vivo experiments, the present study demonstrated that IMD1–53 promoted angiogenesis in primary cultured MMVECs, which was demonstrated by an increase in proliferation, migration and tube formation. In addition, experiments using an in vivo rat model of MI demonstrated that treatment with IMD1–53 significantly increased the capillary density in ischemic myocardium, attenuated LV remodeling and improved post-MI cardiac function, as determined by CD31 staining, Masson trichrome staining and echocardiography, respectively. Furthermore, the addition of Comp C, an AMPK inhibitor, demonstrated that the in vitro and the in vivo effects of IMD1–53 were at least partially dependent on the activation of AMPK. These results indicated that IMD1–53 attenuates post-infarct myocardial remodeling via the promotion of AMPK-dependent angiogenesis. To the best of our knowledge, this is the first demonstration of the involvement of AMPK activation in IMD-induced angiogenesis and is one of only a few studies (8,10,15) to link the cardioprotective role of IMD to therapeutic angiogenesis.

The human IMD gene encodes a prepropeptide of 148 amino acids (preproIMD), which can generate a series of mature peptide fragments by proteolytic cleavage at the C-terminal in vivo (16), such as IMD1–47, IMD8–47 and IMD1–53. Yang et al were the first to report that IMD1–53 stimulated L-Arg transport and increased NOS activity in the rat aorta (15). Furthermore, using direct gene delivery, Smith et al established IMD as a novel angiogenic factor in a rodent hindlimb ischemia model and cultured human umbilical vein endothelial cells (10). Consistent with these findings, the present study demonstrated that IMD1–53 increased proliferation of MMVECs in a dose-dependent manner with a maximal effect at around 80 nmol, and the optimal dose of IMD1–53 treatment promoted migration and tube formation of MMVECs.

Cardiac hypertrophy is initiated as an adaptive response during post-infarction remodeling to compensate for increased load (17). However, the capillary network within the infarcted heart is unable to support the increased demands of the hypertrophied myocardium, resulting in a gradual loss of healthy tissue, extension of the infarct and fibrous replacement (18). Therefore, promotion of angiogenesis in post-infarct myocardium may be an effective approach to attenuate adverse ventricular remodeling following MI, as demonstrated by several previous studies (1921). The present study demonstrated that the in vivo administration of IMD1–53 promoted angiogenesis in post-infarct rat myocardium, which was indicated by increased capillary density in the peri-infarct zone. In addition, chronic treatment with IMD1–53 preserved cardiac geometry and improved cardiac performance post-MI, as evidenced by Masson trichrome staining and echocardiography, suggesting that IMD1–53 may attenuate adverse remodeling post-MI and prevent subsequent heart failure by promoting angiogenesis. IMD1–53 has previously been reported to protect against myocardial ischemia/reperfusion injury (5) and to attenuate pressure-overload-induced cardiac hypertrophy (9) through other mechanisms (6,8); however, to the best of our knowledge, this is the first study to link the cardioprotective role of IMD to therapeutic angiogenesis.

AMPK is a key sensor and regulator of cellular energy homeostasis, which modulates various physiological processes that may favor the restoration of energy balance in several systems (22). In endothelial cells, AMPK was demonstrated to serve an essential role in angiogenesis in response to stress (14,23). In addition, a series of agents, including pravastatin, adiponectin and erythropoietin, and cytokines have been reported to increase angiogenesis through AMPK-dependent mechanisms (2426). Notably, CGRP was also reported to promote angiogenesis by activating the AMPK-eNOS pathway in endothelial cells (27). The present study demonstrated that treatment with IMD1–53 increased the expression levels of p-AMPK in cultured MMVECs and in post-MI rat myocardium, and notably, administration of Comp C, a known inhibitor of AMPK, abrogated the effects of IMD1–53 on angiogenesis, as evidenced by reduced migration and tube formation of MMVECs in vitro, and lower capillary density at the peri-infarct zone in vivo. These results demonstrated that the angiogenic activity of IMD1–53 is at least partially dependent on the activation of AMPK.

It has previously been reported that IMD1–53 increased total NOS activity, but had little effect on mRNA levels of either inducible NOS or eNOS in the rat aorta (16). The results of the current study demonstrated that IMD1–53 promoted the activation of eNOS in vitro and in vivo. In addition, the administration of Comp C significantly attenuated the effects of IMD1–53 on the level of p-eNOS expression, suggesting that AMPK is involved in the IMD1–53-induced activation of eNOS. It is also worth noting that, in the present study, treatment with IMD1–53 had no significant effect on the expression of VEGF in post-infarct rat myocardium, indicating that IMD1–53 may promote the activation of eNOS and subsequent angiogenesis through VEGF-independent mechanisms. Notably, the present study did not determine how IMD1–53 regulated AMPK signaling. In addition, although the α subunit of AMPK was identified as the major target in the present study, further evaluation is required regarding the specific subunit (α1 or α2) involved.

In conclusion, the present study demonstrated that IMD1–53 could attenuate adverse ventricular remodeling post-MI by promoting therapeutic angiogenesis, possibly through the activation of AMPK signaling.

Acknowledgements

The present study was supported by the National Natural Science Foundation of China (grant no. 81100099).

Glossary

Abbreviations

Abbreviations:

Comp C

compound C

FS

fractional shortening

IMD

intermedin

LVEDD

left ventricular end-diastolic diameter

LVEF

left ventricular ejection fraction

LVESD

left ventricular end-systolic diameter; MMVECs;

MI

myocardial infarction

References

1 

Pfeffer MA and Braunwald E: Ventricular remodeling after myocardial infarction. Experimental observations and clinical implications. Circulation. 81:1161–1172. 1990. View Article : Google Scholar : PubMed/NCBI

2 

Jessup M and Brozena S: Heart failure. N Engl J Med. 348:2007–2018. 2003. View Article : Google Scholar : PubMed/NCBI

3 

Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, De Ferranti S, Després JP, Fullerton HJ, Howard VJ, et al: Heart disease and stroke statistics-2015 update: A report from the American Heart Association. Circulation. 131:e29–e322. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Oka T, Akazawa H, Naito AT and Komuro I: Angiogenesis and cardiac hypertrophy: Maintenance of cardiac function and causative roles in heart failure. Circ Res. 114:565–571. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Yang JH, Jia YX, Pan CS, Zhao J, Ouyang M, Yang J, Chang JK, Tang CS and Qi YF: Effects of intermedin(1–53) on cardiac function and ischemia/reperfusion injury in isolated rat hearts. Biochem Biophys Res Commun. 327:713–719. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Zhao L, Peng DQ, Zhang J, Song JQ, Teng X, Yu YR, Tang CS and Qi YF: Extracellular signal-regulated kinase 1/2 activation is involved in intermedin1-53 attenuating myocardial oxidative stress injury induced by ischemia/reperfusion. Peptides. 33:329–335. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Yang X, Zhang H, Jia Y, Ni L, Li G, Xue L and Jiang Y: Effects of intermedin1-53 on myocardial fibrosis. Acta Biochim Biophys Sin (Shanghai). 45:141–148. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Chen H, Wang X, Tong M, Wu D, Wu S, Chen J, Wang X, Wang X, Kang Y, Tang H, et al: Intermedin suppresses pressure overload cardiac hypertrophy through activation of autophagy. PLoS One. 8:e647572013. View Article : Google Scholar : PubMed/NCBI

9 

Lu WW, Zhao L, Zhang JS, Hou YL, Yu YR, Jia MZ, Tang CS and Qi YF: Intermedin1-53 protects against cardiac hypertrophy by inhibiting endoplasmic reticulum stress via activating AMP-activated protein kinase. J Hypertens. 33:1676–1687. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Smith RS Jr, Gao L, Bledsoe G, Chao L and Chao J: Intermedin is a new angiogenic growth factor. Am J Physiol Heart Circ Physiol. 297:H1040–H1047. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Zhang W, Wang LJ, Xiao F, Wei Y, Ke W and Xin HB: Intermedin: A novel regulator for vascular remodeling and tumor vessel normalization by regulating vascular endothelial-cadherin and extracellular signal-regulated kinase. Arterioscler Thromb Vasc Biol. 32:2721–2732. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Derumeaux G, Mulder P, Richard V, Chagraoui A, Nafeh C, Bauer F, Henry JP and Thuillez C: Tissue Doppler imaging differentiates physiological from pathological pressure-overload left ventricular hypertrophy in rats. Circulation. 105:1602–1608. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Wang F, Huang D, Zhu W, Li S, Yan M, Wei M and Li J: Selective inhibition of PKCβ2 preserves cardiac function after myocardial infarction and is associated with improved angiogenesis of ischemic myocardium in diabetic rats. Int J Mol Med. 32:1037–1046. 2013.PubMed/NCBI

14 

Nagata D, Mogi M and Walsh K: AMP-activated protein kinase (AMPK) signaling in endothelial cells is essential for angiogenesis in response to hypoxic stress. J Biol Chem. 278:31000–31006. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Yang JH, Pan CS, Jia YX, Zhang J, Zhao J, Pang YZ, Yang J, Tang CS and Qi YF: Intermedin1-53 activates L-arginine/nitric oxide synthase/nitric oxide pathway in rat aortas. Biochem Biophys Res Commun. 341:567–572. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Roh J, Chang CL, Bhalla A, Klein C and Hsu SY: Intermedin is a calcitonin/calcitonin gene-related peptide family peptide acting through the calcitonin receptor-like receptor/receptor activity-modifying protein receptor complexes. J Biol Chem. 279:7264–7274. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Sutton MG and Sharpe N: Left ventricular remodeling after myocardial infarction: Pathophysiology and therapy. Circulation. 101:2981–2988. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Kocher AA, Schuster MD, Szabolcs MJ, Takuma S, Burkhoff D, Wang J, Homma S, Edwards NM and Itescu S: Neovascularization of ischemic myocardium by human bone-marrow-derived angioblasts prevents cardiomyocyte apoptosis, reduces remodeling and improves cardiac function. Nat Med. 7:430–436. 2001. View Article : Google Scholar : PubMed/NCBI

19 

Li J, Zhang Y, Li C, Xie J, Liu Y, Zhu W, Zhang X, Jiang S, Liu L and Ding Z: HSPA12B attenuates cardiac dysfunction and remodelling after myocardial infarction through an eNOS-dependent mechanism. Cardiovasc Res. 99:674–684. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Vandoorne K, Vandsburger MH, Raz T, Shalev M, Weisinger K, Biton I, Brumfeld V, Raanan C, Nevo N, Eilam R, et al: Chronic Akt1 deficiency attenuates adverse remodeling and enhances angiogenesis after myocardial infarction. Circ Cardiovasc Imaging. 6:992–1000. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Zhao X, Balaji P, Pachon R, Beniamen DM, Vatner DE, Graham RM and Vatner SF: Overexpression of cardiomyocyte α1A-Adrenergic receptors attenuates postinfarct remodeling by inducing angiogenesis through heterocellular signaling. Arterioscler Thromb Vasc Biol. 35:2451–2459. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Grahame Hardie D: AMP-activated protein kinase: A key regulator of energy balance with many roles in human disease. J Intern Med. 276:543–559. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Ahluwalia A and Tarnawski AS: Activation of the metabolic sensor-AMP activated protein kinase reverses impairment of angiogenesis in aging myocardial microvascular endothelial cells. Implications for the aging heart. J Physiol Pharmacol. 62:583–587. 2011.PubMed/NCBI

24 

Izumi Y, Shiota M, Kusakabe H, Hikita Y, Nakao T, Nakamura Y, Muro T, Miura K, Yoshiyama M and Iwao H: Pravastatin accelerates ischemia-induced angiogenesis through AMP-activated protein kinase. Hypertens Res. 32:675–679. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Shimano M, Ouchi N, Shibata R, Ohashi K, Pimentel DR, Murohara T and Walsh K: Adiponectin deficiency exacerbates cardiac dysfunction following pressure overload through disruption of an AMPK-dependent angiogenic response. J Mol Cell Cardiol. 49:210–220. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Su KH, Yu YB, Hou HH, Zhao JF, Kou YR, Cheng LC, Shyue SK and Lee TS: AMP-activated protein kinase mediates erythropoietin-induced activation of endothelial nitric oxide synthase. J Cell Physiol. 227:3053–3062. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Zheng S, Li W, Xu M, Bai X, Zhou Z, Han J, Shyy JY and Wang X: Calcitonin gene-related peptide promotes angiogenesis via AMP-activated protein kinase. Am J Physiol Cell Physiol. 299:C1485–C1492. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2017
Volume 15 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen K, Yan M, Li Y, Dong Z, Huang D, Li J and Wei M: Intermedin1‑53 enhances angiogenesis and attenuates adverse remodeling following myocardial infarction by activating AMP‑activated protein kinase. Mol Med Rep 15: 1497-1506, 2017
APA
Chen, K., Yan, M., Li, Y., Dong, Z., Huang, D., Li, J., & Wei, M. (2017). Intermedin1‑53 enhances angiogenesis and attenuates adverse remodeling following myocardial infarction by activating AMP‑activated protein kinase. Molecular Medicine Reports, 15, 1497-1506. https://doi.org/10.3892/mmr.2017.6193
MLA
Chen, K., Yan, M., Li, Y., Dong, Z., Huang, D., Li, J., Wei, M."Intermedin1‑53 enhances angiogenesis and attenuates adverse remodeling following myocardial infarction by activating AMP‑activated protein kinase". Molecular Medicine Reports 15.4 (2017): 1497-1506.
Chicago
Chen, K., Yan, M., Li, Y., Dong, Z., Huang, D., Li, J., Wei, M."Intermedin1‑53 enhances angiogenesis and attenuates adverse remodeling following myocardial infarction by activating AMP‑activated protein kinase". Molecular Medicine Reports 15, no. 4 (2017): 1497-1506. https://doi.org/10.3892/mmr.2017.6193