NHE3 inhibitor tenapanor maintains intestinal barrier function, decreases visceral hypersensitivity, and attenuates TRPV1 signaling in colonic sensory neurons

Am J Physiol Gastrointest Liver Physiol. 2024 May 1;326(5):G543-G554. doi: 10.1152/ajpgi.00233.2023. Epub 2024 Jan 22.

Abstract

The pathogenesis of irritable bowel syndrome (IBS) is multifactorial, characterized in part by increased intestinal permeability, and visceral hypersensitivity. Increased permeability is associated with IBS severity and abdominal pain. Tenapanor is FDA-approved for the treatment of IBS with constipation (IBS-C) and has demonstrated improvements in bowel motility and a reduction in IBS-related pain; however, the mechanism by which tenapanor mediates these functions remains unclear. Here, the effects of tenapanor on colonic pain signaling and intestinal permeability were assessed through behavioral, electrophysiological, and cell culture experiments. Intestinal motility studies in rats and humans demonstrated that tenapanor increased luminal sodium and water retention and gastrointestinal transit versus placebo. A significantly reduced visceral motor reflex (VMR) to colonic distension was observed with tenapanor treatment versus vehicle in two rat models of visceral hypersensitivity (neonatal acetic acid sensitization and partial restraint stress; both P < 0.05), returning VMR responses to that of nonsensitized controls. Whole cell voltage patch-clamp recordings of retrogradely labeled colonic dorsal root ganglia (DRG) neurons from sensitized rats found that tenapanor significantly reduced DRG neuron hyperexcitability to capsaicin versus vehicle (P < 0.05), an effect not mediated by epithelial cell secretions. Tenapanor also attenuated increases in intestinal permeability in human colon monolayer cultures caused by incubation with proinflammatory cytokines (P < 0.001) or fecal supernatants from patients with IBS-C (P < 0.005). These results support a model in which tenapanor reduces IBS-related pain by strengthening the intestinal barrier, thereby decreasing permeability to macromolecules and antigens and reducing DRG-mediated pain signaling.NEW & NOTEWORTHY A series of nonclinical experiments support the theory that tenapanor inhibits IBS-C-related pain by strengthening the intestinal barrier. Tenapanor treatment reduced visceral motor responses to nonsensitized levels in two rat models of hypersensitivity and reduced responses to capsaicin in sensitized colonic nociceptive dorsal root ganglia neurons. Intestinal permeability experiments in human colon monolayer cultures found that tenapanor attenuates increases in permeability induced by either inflammatory cytokines or fecal supernatants from patients with IBS-C.

Keywords: TRPV1; dorsal root ganglia neurons; intestinal barrier permeability; tenapanor; visceral hypersensitivity.

MeSH terms

  • Abdominal Pain / metabolism
  • Animals
  • Capsaicin / pharmacology
  • Colon / metabolism
  • Cytokines / metabolism
  • Humans
  • Intestinal Barrier Function
  • Irritable Bowel Syndrome* / drug therapy
  • Isoquinolines*
  • Rats
  • Sensory Receptor Cells / metabolism
  • Sodium-Hydrogen Exchanger 3 / metabolism
  • Sulfonamides*
  • TRPV Cation Channels / metabolism

Substances

  • Sodium-Hydrogen Exchanger 3
  • tenapanor
  • Capsaicin
  • Cytokines
  • TRPV1 protein, human
  • TRPV Cation Channels
  • Isoquinolines
  • Sulfonamides

Grants and funding