Protein hydrolysate-induced cholecystokinin secretion from enteroendocrine cells is indirectly mediated by the intestinal oligopeptide transporter PepT1

AP Liou, DI Chavez, E Espero, S Hao… - American Journal …, 2011 - journals.physiology.org
AP Liou, DI Chavez, E Espero, S Hao, SA Wank, HE Raybould
American Journal of Physiology-Gastrointestinal and Liver …, 2011journals.physiology.org
Dietary protein is a major stimulant for cholecystokinin (CCK) secretion by the intestinal I
cell, however, the mechanism by which protein is detected is unknown. Indirect functional
evidence suggests that PepT1 may play a role in CCK-mediated changes in gastric motor
function. However, it is unclear whether this oligopeptide transporter directly or indirectly
activates the I cell. Using both the CCK-expressing enteroendocrine STC-1 cell and acutely
isolated native I cells from CCK-enhanced green fluorescent protein (eGFP) mice, we aimed …
Dietary protein is a major stimulant for cholecystokinin (CCK) secretion by the intestinal I cell, however, the mechanism by which protein is detected is unknown. Indirect functional evidence suggests that PepT1 may play a role in CCK-mediated changes in gastric motor function. However, it is unclear whether this oligopeptide transporter directly or indirectly activates the I cell. Using both the CCK-expressing enteroendocrine STC-1 cell and acutely isolated native I cells from CCK-enhanced green fluorescent protein (eGFP) mice, we aimed to determine whether PepT1 directly activates the enteroendocrine cell to elicit CCK secretion in response to oligopeptides. Both STC-1 cells and isolated CCK-eGFP cells expressed PepT1 transcripts. STC-1 cells were activated, as measured by ERK1/2 phosphorylation, by both peptone and the PepT1 substrate Cefaclor; however, the PepT1 inhibitor 4-aminomethyl benzoic acid (AMBA) had no effect on STC-1 cell activity. The PepT1-transportable substrate glycyl-sarcosine dose-dependently decreased gastric motility in anesthetized rats but had no affect on activation of STC-1 cells or on CCK secretion by CCK-eGFP cells. CCK secretion was significantly increased in response to peptone but not to Cefaclor, cephalexin, or Phe-Ala in CCK-eGFP cells. Taken together, the data suggest that PepT1 does not directly mediate CCK secretion in response to PepT1 specific substrates. PepT1, instead, may have an indirect role in protein sensing in the intestine.
American Physiological Society