Cardiovascular pharmacology
AMP-activated kinase relaxes agonist induced contractions in the mouse aorta via effects on PKC signaling and inhibits NO-induced relaxation

https://doi.org/10.1016/j.ejphar.2012.07.025Get rights and content

Abstract

Adenosine monophosphate activated kinase (AMPK), a regulator of cellular metabolism, has been shown to relax arterial smooth muscle via endothelium-dependent and independent mechanisms. We have examined the role of AMPK in different smooth muscles using the activating compound, 5-amino-4-imidazolecarboxamide riboside-1-β-d-ribofuranoside (AICAR). Isolated preparations of mouse aorta, saphenous artery, ileum and urinary bladder were compared. AICAR produced a reversible dose-dependent relaxation in aortic rings pre-incubated with AICAR and activated with phenylephrine. Less prominent relaxation was noted in the other tissues. This difference in sensitivity to AICAR was not due to differences in the expression levels of AMPK α1 mRNA. In the aorta, AICAR had a greater effect on contractions induced by phenylephrine, compared to high-K+ induced contractions. Contractions of the aorta in response to the protein kinase C activator PDBu were prominently inhibited by AICAR. The AICAR relaxation observed in the aorta was not prevented by the NOS inhibitor L-NAME, Indomethacin or endothelium removal. Nitric oxide (NO) mediated relaxations in aortic preparations induced by acetylcholine or sodium nitroprusside (SNP) were attenuated by AICAR. In conclusion, AMPK induced relaxation of smooth muscle is tissue-dependent and most prominent in large elastic arteries. The smooth muscle relaxation is NO-independent and occurs downstream of PKC activation and is associated with attenuated relaxant responses to NO.

Introduction

Adenosine-monophosphate-activated-kinase (AMPK) is a widely expressed serine-threonine protein kinase that functions as a cellular energy sensor (Hardie, 2011). It consists of a catalytic α subunit, expressed as α1 and α2 isoforms, and regulatory (β, γ) subunits. It is activated by phosphorylation by kinases primarily acting on a threonine-residue on the α-subunit (Hawley et al., 1996). This phosphorylation is favored by AMP interaction, which also opposes dephosphorylation and inactivation (Cheung et al., 2000). These effects of AMP are antagonized by ATP, thus rendering AMPK activity responsive to changing [AMP]/[ATP] ratios. AMPK has numerous targets in pathways involved in energy metabolism and growth regulation and several proposed physiological functions. In general, AMPK activation results in inhibition of ATP-consuming, anabolic, metabolic pathways, while stimulating pathways that generate ATP (Hardie, 2011). In addition to the physiological activators of AMPK, a range of pharmacological compounds, including the nucleoside compound 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR), activate the enzyme (Corton et al., 1995).

In skeletal muscle, AMPK has a role in glucose uptake and has been proposed as a potential target for treating insulin resistance (Kraegen et al., 2009). The detailed functions of AMPK in smooth muscle tissue are largely unknown. AMPK is expressed in smooth muscle and in endothelial cells (Dagher et al., 1999, Rubin et al., 2005). In rat pulmonary arteries, AMPK is activated during hypoxia (Robertson et al., 2008) and AMPK is suggested to couple altered mitochondrial respiration to hypoxic pulmonary vasoconstriction (Evans, 2006). In contrast, AMPK activation is associated with vasodilation in systemic vascular beds. AMPK activates eNOS in vascular endothelium during hypoxia (Nagata and Hirata, 2010). AMPK dilates rat resistance arteries (Bradley et al., 2010) and has been reported to lower blood pressure in vivo (Foley et al., 1989, Bosselaar et al., 2011). In spontaneously hypertensive rats, AMPK activation causes endothelium-NO-dependent vasorelaxation (Ford and Rush, 2011, Ford et al., 2012). In a more long-term perspective, AMPK inhibits proliferation and hypertrophy of vascular smooth muscle (Nagata et al., 2004, Igata et al., 2005, Ferri, 2012). AMPK also has been shown to relax vascular smooth muscle in mouse aorta independently of the endothelium (Goirand et al., 2007). Mechanisms for this effect are unknown and direct actions on the myosin light chain kinase (Horman et al., 2008) or small G protein signaling (Wang et al., 2011) may be involved.

The effects of AMPK activation, and AICAR, have been primarily studied in isolated arteries activated with receptor agonists. Information on other smooth muscle tissues and different modes of activation is not available. The relative contribution of endothelial effects and effects from direct action on smooth muscle are not resolved and the mechanisms for the relaxant effects in smooth muscle are unknown. We have therefore addressed the following questions: (1) Is the relaxant effect of AICAR also observed in a broader range of smooth muscle tissues?, (2) Does AICAR relax smooth muscle via effects on receptor activation or on downstream cellular signaling pathways?, and (3) What are the effects of AICAR on endothelial mediated relaxation in the arteries?

Section snippets

Animals and preparations

Adult female C57/Bl6 mice (approximately 8–12 weeks old from Taconic A/S, Denmark or B&K Universal AB, Sweden) were euthanized by cervical dislocation, and the urinary bladder, aorta, ileum and saphenous artery (in the hind limb, distal to the femoral artery) were rapidly removed and further dissected in cold Krebs-Ringer solution (for composition see Section 2.2). All experiments were approved by the local animal ethics committee and conformed to the European guidelines for animal research.

Solutions and chemicals

The

Effects of AICAR on contractions induced by depolarization and agonists in different smooth muscle tissues

Fig. 1, Panel A summarizes the effects of AICAR on contractions induced by high-K+ (80 mM) in different smooth muscle tissues. Only minor effects of AICAR were observed, except at the highest examined dose (1 mM) where the force of the ileum preparations was inhibited by about 35% compared to the response in the absence of the compound (triangles base up) (control ileum: 0.84±0.08; AICAR: 0.54±0.05; P<0.05, unpaired Student's t test, responses relative to an initial 80 mM KCl-induced contraction).

Discussion

We show that AICAR relaxes smooth muscle in a tissue-dependent manner, with aortic vascular muscle exhibiting a greater sensitivity than the other tissues investigated. The selectivity of AICAR in the vascular tree for the large elastic artery compared to the resistance sized artery suggests that AMPK can constitute a mechanism for metabolic regulation of tone in the conduit arteries. This pathway might be active in altering arterial compliance during metabolic stress. The relaxant effects of

Acknowledgements

This study was supported by the Swedish Research Council, the Karolinska Institutet, an FP7 EU grant (INComb), and the Swedish Heart-Lung Foundation.

References (37)

  • A. Arner et al.

    Smooth, slow and smart muscle motors

    J. Muscle Res. Cell Motil.

    (2003)
  • L. Boberg et al.

    Role of Rho-kinase and protein kinase C during contraction of hypertrophic detrusor in mice with partial urinary bladder outlet obstruction

    BJU Int.

    (2012)
  • M. Bosselaar et al.

    Intravenous AICAR during hyperinsulinemia induces systemic hemodynamic changes but has no local metabolic effect

    J. Clin. Pharmacol.

    (2011)
  • E.A. Bradley et al.

    Activation of AMP-activated protein kinase by 5-aminoimidazole-4-carboxamide-1-beta-d-ribofuranoside in the muscle microcirculation increases nitric oxide synthesis and microvascular perfusion

    Arteriosclerosis Thromb. Vasc. Biol.

    (2010)
  • J.M. Cacicedo et al.

    Acute exercise activates AMPK and eNOS in the mouse aorta

    Am. J. Physiol. Heart Circ. Physiol.

    (2011)
  • T.-C. Chang et al.

    Lin-28B transactivation is necessary for Myc-mediated let-7 repression and proliferation

    PNAS

    (2009)
  • P.C. Cheung et al.

    Characterization of AMP-activated protein kinase gamma-subunit isoforms and their role in AMP binding

    Biochem. J.

    (2000)
  • J.M. Corton et al.

    5-aminoimidazole-4-carboxamide ribonucleoside. A specific method for activating AMP-activated protein kinase in intact cells?

    Eur. J. Biochem.

    (1995)
  • Cited by (14)

    • Cellular and molecular biology of aging endothelial cells

      2015, Journal of Molecular and Cellular Cardiology
      Citation Excerpt :

      However, despite evidence for AMPK-mediated eNOS activation [238,240–242], the effect of in vivo AMPK activation by AICAR to improve dilation in arteries from old mice was not mediated by an increased NO bioavailability [247]. Similar NO independent effects of AICAR were also found after acute in vitro administration to isolated aortic rings, in which AICAR induced relaxation of agonist constricted vessels independent of NO [249]. Similar to treatment of old mice with SIRT-1 activators, AMPK activators therefore reduce arterial oxidative stress and improve endothelial function, but do so via an NO independent mechanism.

    • Novel therapeutic strategies for adult obese asthmatics

      2014, Immunology and Allergy Clinics of North America
      Citation Excerpt :

      Metformin can also increase NO synthase 3 (NOS3) dependent production of NO and improve endothelial function through AMPK-dependent positive regulation of NOS3 activity and inhibition of NOS3 negative regulators. Treatment of endothelial cell lines and mice with metformin increases AMPK-dependent NOS3 phosphorylation at the regulatory site Ser1177/1179 and NO production.12–14 NOS3 activity is negatively regulated through phosphorylation of Thr495 by protein kinase C-β (PKCβ), which is up-regulated in human asthmatics and patients with insulin resistance.15

    View all citing articles on Scopus
    View full text