Abstract

The large conductance voltage- and Ca(2+)-activated K(+) channel (MaxiK, BK(Ca), BK) is composed of four pore-forming α-subunits and can be associated with regulatory β-subunits. One of the functional roles of MaxiK is to regulate vascular tone. We recently found that the MaxiK channel from coronary smooth muscle is trans-inhibited by activation of the vasoconstricting thromboxane A(2) prostanoid receptor (TP), a mechanism supported by MaxiK α-subunit (MaxiKα)-TP physical interaction. Here, we examined the role of the MaxiK β1-subunit in TP-MaxiK association. We found that the β1-subunit can by itself interact with TP and that this association can occur independently of MaxiKα. Subcellular localization analysis revealed that β1 and TP are closely associated at the cell periphery. The molecular mechanism of β1-TP interaction involves predominantly the β1 extracellular loop. As reported previously, TP activation by the thromboxane A(2) analog U46619 caused inhibition of MaxiKα macroscopic conductance or fractional open probability (FP(o)) as a function of voltage. However, the positive shift of the FP(o) versus voltage curve by U46619 relative to the control was less prominent when β1 was coexpressed with TP and MaxiKα proteins (20 ± 6 mV, n = 7) than in cells expressing TP and MaxiKα alone (51 ± 7 mV, n = 7). Finally, β1 gene ablation reduced the EC(50) of the U46619 agonist in mediating aortic contraction from 18 ± 1 nm (n = 12) to 9 ± 1 nm (n = 12). The results indicate that the β1-subunit can form a tripartite complex with TP and MaxiKα, has the ability to associate with each protein independently, and diminishes U46619-induced MaxiK channel trans-inhibition as well as vasoconstriction.

Highlights

  • The vasoconstricting thromboxane A2 prostanoid receptor (TP) is physically coupled to the MaxiK channel ␣-subunit, down-regulating its activity, but the role of the MaxiK ␤1-subunit is unknown

  • We have recently shown that activation of TP trans-inhibits MaxiK channel activity in native coronary arterial muscle; this G-protein independent coupling is supported by the ability of TP to physically interact with the MaxiK ␣-subunit (MaxiK␣) (12)

  • The results from this study reveal the following. (i) The MaxiK regulatory ␤1-subunit can by itself interact with vasoconstricting TP. (ii) ␤1 expression decreases activated TP-induced positive shift in MaxiK␣ voltage dependence of the fractional open probability. (iii) ␤1 expression is linked to a reduced potency of the thromboxane A2 analog U46619 in producing vasoconstriction

Read more

Summary

Background

The vasoconstricting thromboxane A2 prostanoid receptor (TP) is physically coupled to the MaxiK channel ␣-subunit, down-regulating its activity, but the role of the MaxiK ␤1-subunit is unknown. We recently found that the MaxiK channel from coronary smooth muscle is trans-inhibited by activation of the vasoconstricting thromboxane A2 prostanoid receptor (TP), a mechanism supported by MaxiK ␣-subunit (MaxiK␣)-TP physical interaction. A few studies have examined the role of ␤1 in vasorelaxant- or vasoconstrictor-mediated alterations of MaxiK channel activity In this respect, ␤1 is essential for lithocholatemediated MaxiK activation linked to cerebral vasodilation and for alcohol-induced MaxiK inhibition and associated vasoconstriction (10, 11), yet the ␤1 relationship to vasoconstricting G protein-coupled receptors such as the thromboxane A2 prostanoid receptor (TP) is unknown. We have recently shown that activation of TP trans-inhibits MaxiK channel activity in native coronary arterial muscle; this G-protein independent coupling is supported by the ability of TP to physically interact with the MaxiK ␣-subunit (MaxiK␣) (12). Because in coronary arterial muscle the majority of currents resemble channels accompanied by the ␤1-subunit (13), the question of whether (and how) ␤1 relates to vasoconstricting G protein-coupled receptors (in this case) to TP actions becomes relevant

The abbreviations used are
EXPERIMENTAL PROCEDURES
RESULTS
A N-terminus
Findings
DISCUSSION
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call