Promiscuous Dimerization of the Growth Hormone Secretagogue Receptor (GHS-R1a) Attenuates Ghrelin-mediated Signaling

Typeset version

 

TY  - JOUR
  - Schellekens, H,van Oeffelen, WEPA,Dinan, TG,Cryan, JF
  - 2013
  - January
  - The Journal of Biological Chemistry
  - Promiscuous Dimerization of the Growth Hormone Secretagogue Receptor (GHS-R1a) Attenuates Ghrelin-mediated Signaling
  - Validated
  - ()
  - PROTEIN-COUPLED RECEPTORS MESSENGER-RNA CONSTITUTIVE ACTIVITY LENTIVIRAL-VECTORS NONDIVIDING CELLS GENE-TRANSFER HIGH-AFFINITY FOOD REWARD CROSS-TALK SEROTONIN
  - 288
  - 181
  - 191
  - G protein-coupled receptors (GPCRs), such as the ghrelin receptor (GHS-R1a), the melanocortin 3 receptor (MC3), and the serotonin 2C receptor (5-HT2C), are well known for their key role in the homeostatic control of food intake and energy balance. Ghrelin is the only known gut peptide exerting an orexigenic effect and has thus received much attention as an antiobesity drug target. In addition, recent data have revealed a critical role for ghrelin in dopaminergic mesolimbic circuits involved in food reward signaling. This study investigates the downstream signaling consequences and ligand-mediated co-internalization following heterodimerization of the GHS-R1a receptor with the dopamine 1 receptor, as well as that of the GHS-R1a-MC3 heterodimer. In addition, a novel heterodimer between the GHS-R1a receptor and the 5-HT2C receptor was identified. Interestingly, dimerization of the GHS-R1a receptor with the unedited 5-HT2C-INI receptor, but not with the partially edited 5-HT2C-VSV isoform, significantly reduced GHS-R1a agonist-mediated calcium influx, which was completely restored following pharmacological blockade of the 5-HT2C receptor. These results combined suggest a potential novel mechanism for fine-tuning GHS-R1a receptor-mediated activity via promiscuous dimerization of the GHS-R1a receptor with other G protein-coupled receptors involved in appetite regulation and food reward. These findings may uncover novel mechanisms of significant relevance for the future pharmacological targeting of the GHS-R1a receptor in the homeostatic regulation of energy balance and in hedonic appetite signaling, both of which play a significant role in the development of obesity.
  - DOI 10.1074/jbc.M112.382473
DA  - 2013/01
ER  - 
@article{V206308059,
   = {Schellekens,  H and van Oeffelen,  WEPA and Dinan,  TG and Cryan,  JF },
   = {2013},
   = {January},
   = {The Journal of Biological Chemistry},
   = {Promiscuous Dimerization of the Growth Hormone Secretagogue Receptor (GHS-R1a) Attenuates Ghrelin-mediated Signaling},
   = {Validated},
   = {()},
   = {PROTEIN-COUPLED RECEPTORS MESSENGER-RNA CONSTITUTIVE ACTIVITY LENTIVIRAL-VECTORS NONDIVIDING CELLS GENE-TRANSFER HIGH-AFFINITY FOOD REWARD CROSS-TALK SEROTONIN},
   = {288},
  pages = {181--191},
   = {{G protein-coupled receptors (GPCRs), such as the ghrelin receptor (GHS-R1a), the melanocortin 3 receptor (MC3), and the serotonin 2C receptor (5-HT2C), are well known for their key role in the homeostatic control of food intake and energy balance. Ghrelin is the only known gut peptide exerting an orexigenic effect and has thus received much attention as an antiobesity drug target. In addition, recent data have revealed a critical role for ghrelin in dopaminergic mesolimbic circuits involved in food reward signaling. This study investigates the downstream signaling consequences and ligand-mediated co-internalization following heterodimerization of the GHS-R1a receptor with the dopamine 1 receptor, as well as that of the GHS-R1a-MC3 heterodimer. In addition, a novel heterodimer between the GHS-R1a receptor and the 5-HT2C receptor was identified. Interestingly, dimerization of the GHS-R1a receptor with the unedited 5-HT2C-INI receptor, but not with the partially edited 5-HT2C-VSV isoform, significantly reduced GHS-R1a agonist-mediated calcium influx, which was completely restored following pharmacological blockade of the 5-HT2C receptor. These results combined suggest a potential novel mechanism for fine-tuning GHS-R1a receptor-mediated activity via promiscuous dimerization of the GHS-R1a receptor with other G protein-coupled receptors involved in appetite regulation and food reward. These findings may uncover novel mechanisms of significant relevance for the future pharmacological targeting of the GHS-R1a receptor in the homeostatic regulation of energy balance and in hedonic appetite signaling, both of which play a significant role in the development of obesity.}},
   = {DOI 10.1074/jbc.M112.382473},
  source = {IRIS}
}
AUTHORSSchellekens, H,van Oeffelen, WEPA,Dinan, TG,Cryan, JF
YEAR2013
MONTHJanuary
JOURNAL_CODEThe Journal of Biological Chemistry
TITLEPromiscuous Dimerization of the Growth Hormone Secretagogue Receptor (GHS-R1a) Attenuates Ghrelin-mediated Signaling
STATUSValidated
TIMES_CITED()
SEARCH_KEYWORDPROTEIN-COUPLED RECEPTORS MESSENGER-RNA CONSTITUTIVE ACTIVITY LENTIVIRAL-VECTORS NONDIVIDING CELLS GENE-TRANSFER HIGH-AFFINITY FOOD REWARD CROSS-TALK SEROTONIN
VOLUME288
ISSUE
START_PAGE181
END_PAGE191
ABSTRACTG protein-coupled receptors (GPCRs), such as the ghrelin receptor (GHS-R1a), the melanocortin 3 receptor (MC3), and the serotonin 2C receptor (5-HT2C), are well known for their key role in the homeostatic control of food intake and energy balance. Ghrelin is the only known gut peptide exerting an orexigenic effect and has thus received much attention as an antiobesity drug target. In addition, recent data have revealed a critical role for ghrelin in dopaminergic mesolimbic circuits involved in food reward signaling. This study investigates the downstream signaling consequences and ligand-mediated co-internalization following heterodimerization of the GHS-R1a receptor with the dopamine 1 receptor, as well as that of the GHS-R1a-MC3 heterodimer. In addition, a novel heterodimer between the GHS-R1a receptor and the 5-HT2C receptor was identified. Interestingly, dimerization of the GHS-R1a receptor with the unedited 5-HT2C-INI receptor, but not with the partially edited 5-HT2C-VSV isoform, significantly reduced GHS-R1a agonist-mediated calcium influx, which was completely restored following pharmacological blockade of the 5-HT2C receptor. These results combined suggest a potential novel mechanism for fine-tuning GHS-R1a receptor-mediated activity via promiscuous dimerization of the GHS-R1a receptor with other G protein-coupled receptors involved in appetite regulation and food reward. These findings may uncover novel mechanisms of significant relevance for the future pharmacological targeting of the GHS-R1a receptor in the homeostatic regulation of energy balance and in hedonic appetite signaling, both of which play a significant role in the development of obesity.
PUBLISHER_LOCATION
ISBN_ISSN
EDITION
URL
DOI_LINKDOI 10.1074/jbc.M112.382473
FUNDING_BODY
GRANT_DETAILS