Dipeptidyl peptidase IV inhibition potentiates amino acid- and bile acid-induced bicarbonate secretion in rat duodenum

  • Takuya Inoue
    Department of Medicine, School of Medicine, University of California, Los Angeles, California;
  • Joon-Ho Wang
    Department of Medicine, School of Medicine, University of California, Los Angeles, California;
  • Masaaki Higashiyama
    Department of Medicine, School of Medicine, University of California, Los Angeles, California;
  • Sergiy Rudenkyy
    Greater Los Angeles Veterans Affairs Healthcare System, University of California, Los Angeles, California;
  • Kazuhide Higuchi
    The Second Department of Internal Medicine, Osaka Medical College, Osaka, Japan
  • Paul H. Guth
    Greater Los Angeles Veterans Affairs Healthcare System, University of California, Los Angeles, California;
  • Eli Engel
    Department of Biomathematics, University of California, Los Angeles, California;
  • Jonathan D. Kaunitz
    Greater Los Angeles Veterans Affairs Healthcare System, University of California, Los Angeles, California;
  • Yasutada Akiba
    Greater Los Angeles Veterans Affairs Healthcare System, University of California, Los Angeles, California;

抄録

<jats:p> Intestinal endocrine cells release gut hormones, including glucagon-like peptides (GLPs), in response to luminal nutrients. Luminal l-glutamate (l-Glu) and 5′-inosine monophosphate (IMP) synergistically increases duodenal HCO<jats:sub>3</jats:sub><jats:sup>−</jats:sup> secretion via GLP-2 release. Since L cells express the bile acid receptor TGR5 and dipeptidyl peptidase (DPP) IV rapidly degrades GLPs, we hypothesized that luminal amino acids or bile acids stimulate duodenal HCO<jats:sub>3</jats:sub><jats:sup>−</jats:sup> secretion via GLP-2 release, which is enhanced by DPPIV inhibition. We measured HCO<jats:sub>3</jats:sub><jats:sup>−</jats:sup> secretion with pH and CO<jats:sub>2</jats:sub> electrodes using a perfused rat duodenal loop under isoflurane anesthesia. l-Glu (10 mM) and IMP (0.1 mM) were luminally coperfused with or without luminal perfusion (0.1 mM) or intravenous (iv) injection (3 μmol/kg) of the DPPIV inhibitor NVP728. The loop was also perfused with a selective TGR5 agonist betulinic acid (BTA, 10 μM) or the non-bile acid type TGR5 agonist 3-(2-chlorophenyl)- N-(4-chlorophenyl)- N,5-dimethylisoxazole-4-carboxamide (CCDC; 10 μM). DPPIV activity visualized by use of the fluorogenic substrate was present on the duodenal brush border and submucosal layer, both abolished by the incubation with NVP728 (0.1 mM). An iv injection of NVP728 enhanced l-Glu/IMP-induced HCO<jats:sub>3</jats:sub><jats:sup>−</jats:sup> secretion, whereas luminal perfusion of NVP728 had no effect. BTA or CCDC had little effect on HCO<jats:sub>3</jats:sub><jats:sup>−</jats:sup> secretion, whereas NVP728 iv markedly enhanced BTA- or CCDC-induced HCO<jats:sub>3</jats:sub><jats:sup>−</jats:sup> secretion, the effects inhibited by a GLP-2 receptor antagonist. Coperfusion of the TGR5 agonist enhanced l-Glu/IMP-induced HCO<jats:sub>3</jats:sub><jats:sup>−</jats:sup> secretion with the enhanced GLP-2 release, suggesting that TGR5 activation amplifies nutrient sensing signals. DPPIV inhibition potentiated luminal l-Glu/IMP-induced and TGR5 agonist-induced HCO<jats:sub>3</jats:sub><jats:sup>−</jats:sup> secretion via a GLP-2 pathway, suggesting that the modulation of the local concentration of the endogenous secretagogue GLP-2 by luminal compounds and DPPIV inhibition helps regulate protective duodenal HCO<jats:sub>3</jats:sub><jats:sup>−</jats:sup> secretion. </jats:p>

収録刊行物

被引用文献 (6)*注記

もっと見る

詳細情報 詳細情報について

問題の指摘

ページトップへ