Quantification of dynamic protein complexes using Renilla luciferase fragment complementation applied to protein kinase A activities in vivo

E Stefan, S Aquin, N Berger… - Proceedings of the …, 2007 - National Acad Sciences
E Stefan, S Aquin, N Berger, CR Landry, B Nyfeler, M Bouvier, SW Michnick
Proceedings of the National Academy of Sciences, 2007National Acad Sciences
The G protein-coupled receptor (GPCR) superfamily represents the most important class of
pharmaceutical targets. Therefore, the characterization of receptor cascades and their
ligands is a prerequisite to discovering novel drugs. Quantification of agonist-induced
second messengers and downstream-coupled kinase activities is central to characterization
of GPCRs or other pathways that converge on GPCR-mediated signaling. Furthermore,
there is a need for simple, cell-based assays that would report on direct or indirect actions …
The G protein-coupled receptor (GPCR) superfamily represents the most important class of pharmaceutical targets. Therefore, the characterization of receptor cascades and their ligands is a prerequisite to discovering novel drugs. Quantification of agonist-induced second messengers and downstream-coupled kinase activities is central to characterization of GPCRs or other pathways that converge on GPCR-mediated signaling. Furthermore, there is a need for simple, cell-based assays that would report on direct or indirect actions on GPCR-mediated effectors of signaling. More generally, there is a demand for sensitive assays to quantify alterations of protein complexes in vivo. We describe the development of a Renilla luciferase (Rluc)-based protein fragment complementation assay (PCA) that was designed specifically to investigate dynamic protein complexes. We demonstrate these features for GPCR-induced disassembly of protein kinase A (PKA) regulatory and catalytic subunits, a key effector of GPCR signaling. Taken together, our observations show that the PCA allows for direct and accurate measurements of live changes of absolute values of protein complex assembly and disassembly as well as cellular imaging and dynamic localization of protein complexes. Moreover, the Rluc-PCA has a sufficiently high signal-to-background ratio to identify endogenously expressed Gαs protein-coupled receptors. We provide pharmacological evidence that the phosphodiesterase-4 family selectively down-regulates constitutive β-2 adrenergic- but not vasopressin-2 receptor-mediated PKA activities. Our results show that the sensitivity of the Rluc-PCA simplifies the recording of pharmacological profiles of GPCR-based candidate drugs and could be extended to high-throughput screens to identify novel direct modulators of PKA or upstream components of GPCR signaling cascades.
National Acad Sciences