Archives

  • 2018-07
  • 2019-04
  • 2019-05
  • 2019-06
  • 2019-07
  • 2019-08
  • 2019-09
  • 2019-10
  • 2019-11
  • 2019-12
  • 2020-01
  • 2020-02
  • 2020-03
  • 2020-04
  • 2020-05
  • 2020-06
  • 2020-07
  • 2020-08
  • 2020-09
  • 2020-10
  • 2020-11
  • 2020-12
  • 2021-01
  • 2021-02
  • 2021-03
  • 2021-04
  • 2021-05
  • 2021-06
  • 2021-07
  • 2021-08
  • 2021-09
  • 2021-10
  • 2021-11
  • 2021-12
  • 2022-01
  • 2022-02
  • 2022-03
  • 2022-04
  • 2022-05
  • 2022-06
  • 2022-07
  • 2022-08
  • 2022-09
  • 2022-10
  • 2022-11
  • 2022-12
  • 2023-01
  • 2023-02
  • 2023-03
  • 2023-04
  • 2023-05
  • 2023-06
  • 2023-07
  • 2023-08
  • 2023-09
  • 2023-10
  • 2023-11
  • 2023-12
  • 2024-01
  • 2024-02
  • 2024-03
  • 2024-04
  • Acknowledgments We would like to

    2022-05-18

    Acknowledgments We would like to thank cannabinoid receptor agonist Dr. Greg Morriello, Mr. Lehua Chang, Mr. Chris Moyes and Mr. Nam Fung Kar for intermediate 3 syntheses. Cheng Zhu would like to thank Dr. Milana Maletic, Mr. Kake Zhao and Mrs. Wanying Sun for helpful discussions.
    Introduction Glucagon-like peptide-1 (GLP-1) has multiple anti-diabetic effects, most notably enhancing insulin secretion, suppressing glucagon release and slowing gastric emptying [1]. Current incretin-based therapies focus on preventing the breakdown of GLP-1 by dipeptidyl peptidase-IV or administrating GLP-1 mimetics [2]. The benefits of increasing endogenous GLP-1 secretion are currently under evaluation, supported by evidence that gastric bypass surgery improves glucose tolerance, at least in part by increased GLP-1 secretion [3]. GPR119 is one of a number of candidate G-protein coupled receptors currently under investigation as a potential target for elevating GLP-1 and insulin release. GLP-1 is secreted from enteroendocrine L-cells in the intestinal epithelium, which express a variety of receptors and transporters capable of detecting ingested nutrients, including carbohydrates, lipids and proteins [4]. GPR119 is a Gαs-coupled receptor, linked to the elevation of intracellular cAMP concentrations [5], [6], [7], [8], [9], [10], [11]. Physiological GPR119 ligands include oleoylethanolamide (OEA) [6], produced locally within tissues [12], [13], [14], and 2-oleoyl glycerol (2-OG) [15] generated by luminal triacylglycerol cannabinoid receptor agonist [16]. OEA as well as small molecule GPR119 agonists, increase GLP-1 and insulin release in rodent models [9], [17], [18], [19]. Indeed, GPR119 agonists were developed for human studies and taken into clinical trials in patients with type 2 diabetes, but were not found to improve metabolic control [20]. The reasons for the poor translatability remain uncertain, and the physiological roles and therapeutic potential of GPR119 are still under investigation.
    Methods
    Results
    Discussion Following the de-orphanization of GPR119, small molecules targeting this receptor were developed as potential new treatments for diabetes that would increase secretion from intestinal L-cells [25]. Although subsequent trials have not yet demonstrated that metabolic improvements can be brought about by the use of GPR119 agonists in humans with type 2 diabetes [20], there is still a high level of academic and commercial interest in GPR119 as a potential drug target [26], [27]. Our results show that L-cell GPR119 is a critical component of the sensing mechanism responsible for GLP-1 responses to ingested lipid, and that L-cells in the distal intestine respond to GPR119 agonists with elevated cAMP and GLP-1 secretion. We show here that GPR119 ligands increase GLP-1 release from primary cultured ileal and colonic L-cells in a GPR119-dependent manner. Of the three GPR119 agonists tested, OEA and AR231453 were more effective than 2-OG. The magnitude of the secretory response triggered by the different GPR119 ligands increased progressively from the upper small intestine to the colon. Indeed, L-cell knockout of Gpr119 largely abolished responses to OEA, 2-OG and AR231453 in the colon. In the ileum, where the secretory response was smaller, only OEA and AR231453 raised secretion in WT tissues above that found in the Gpr119-KO, and in the duodenum/jejunum, none of the ligands had a greater effect in WT than KO cultures. While our results suggest that the small response to OEA in the duodenum/jejunum of WT tissue is independent of GPR119, we cannot exclude the possibility that the proportion of L-cells undergoing Cre-dependent GPR119 excision differed between tissues and that more residual L-cells expressed GPR119 in the upper intestine. Arguing against this idea, however, AR231453 had little effect on GLP-1 secretion in the WT duodenum/jejunum, and OEA has been reported to activate other pathways such as PPARα that might influence GLP-1 secretion even in the absence of Gpr119 [28].