Explaining divergent observations regarding osteocalcin/GPRC6A endocrine signaling

M Pi, SK Nishimoto, L Darryl Quarles - Endocrinology, 2021 - academic.oup.com
M Pi, SK Nishimoto, L Darryl Quarles
Endocrinology, 2021academic.oup.com
A new schema proposes that the bone-derived osteocalcin (Ocn) peptide hormone activates
the G-protein–coupled receptor GPRC6A to directly regulate glucose and fat metabolism in
liver, muscle, and fat, and to stimulate the release of metabolism-regulating hormones,
including insulin, fibroblast growth factor 21, glucagon-like peptide 1, testosterone, and
interleukin 6. Ocn/GPRC6A activation has also been implicated in cancer progression.
GPRC6A is activated by cations, amino acids, and testosterone. The multiligand specificity …
Abstract
A new schema proposes that the bone-derived osteocalcin (Ocn) peptide hormone activates the G-protein–coupled receptor GPRC6A to directly regulate glucose and fat metabolism in liver, muscle, and fat, and to stimulate the release of metabolism-regulating hormones, including insulin, fibroblast growth factor 21, glucagon-like peptide 1, testosterone, and interleukin 6. Ocn/GPRC6A activation has also been implicated in cancer progression. GPRC6A is activated by cations, amino acids, and testosterone. The multiligand specificity, the regulation of energy metabolism in diverse tissues, and the coordinated release of metabolically active hormones make the GPRC6A endocrine networks unique. Recently, the significance of Ocn/GPRCA has been questioned. There is a lack of metabolic abnormalities in newly created genetically engineered Ocn- and Gprc6a-deficient mouse models. There are also paradoxical observations that GPRC6A may function as a tumor suppressor. In addition, discordant published studies have cast doubt on the function of the most prevalent uniquely human GPRC6A-KGKY polymorphism. Explanations for these divergent findings are elusive. We provide evidence that the metabolic susceptibility of genetically engineered Ocn- and Gprc6a-deficient mice is influenced by environmental challenges and genetic differences in mouse strains. In addition, the GPRC6A-KGKY polymorphism appears to be a gain-of-function variant. Finally, alternatively spliced isoforms of GPRC6A may alter ligand specificity and signaling that modulate oncogenic effects. Thus, genetic, post-translational and environmental factors likely account for the variable results regarding the functions of GPRC6A in animal models. Pending additional information, GPRC6A should remain a potential therapeutic target for regulating energy and fat metabolism, hormone production, and cancer progression.
Oxford University Press