Intestinal alkaline phosphatase inhibits the translocation of bacteria of gut-origin in mice with peritonitis: mechanism of action

PLoS One. 2015 May 6;10(5):e0124835. doi: 10.1371/journal.pone.0124835. eCollection 2015.

Abstract

Exogenous intestinal alkaline phosphatase (IAP), an enzyme produced endogenously at the brush edge of the intestinal mucosa, may mitigate the increase in aberrant intestinal permeability increased during sepsis. The aim of this study was to test the efficacy of the inhibitory effect of IAP on acute intestinal inflammation and to study the molecular mechanisms underlying IAP in ameliorating intestinal permeability. We used an in vivo imaging method to evaluate disease status and the curative effect of IAP. Two Escherichia coli (E.coli) B21 strains, carrying EGFP labeled enhanced green fluorescent protein (EGFP) and RFP labeled red fluorescent protein (RFP), were constructed as tracer bacteria and were administered orally to C57/B6N mice to generate an injection peritonitis (IP) model. The IP model was established by injecting inflammatory lavage fluid. C57/B6N mice bearing the tracer bacteria were subsequently treated with (IP+IAP group), or without IAP (IP group). IAP was administered to the mice via tail vein injections. The amount of tracer bacteria in the blood, liver, and lungs at 24 h post-injection was analyzed via flow cytometry (FCM), in vivo imaging, and Western blotting. Intestinal barrier function was measured using a flux assay with the macro-molecule fluorescein isothiocyanate dextran, molecular weight 40kD, (FD40). To elucidate the molecular mechanism underlying the effects of IAP, we examined the levels of ERK phosphorylation, and the expression levels of proteins in the ERK-SP1-VEGF and ERK-Cdx-2-Claudin-2 pathways. We observed that IAP inhibited the expression of Claudin-2, a type of cation channel-forming protein, and VEGF, a cytokine that may increase intestinal permeability by reducing the levels of dephosphorylated ERK. In conclusion, exogenous IAP shows a therapeutic effect in an injection peritonitis model. This including inhibition of bacterial translocation. Moreover, we have established an imaging methodology for live-animals can effectively evaluate intestinal permeability and aberrant bacterial translocation in IP models.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Alkaline Phosphatase / administration & dosage*
  • Animals
  • Blood / microbiology
  • Caco-2 Cells
  • Disease Models, Animal
  • Escherichia coli / chemistry*
  • Escherichia coli / drug effects*
  • Escherichia coli / physiology
  • Extracellular Signal-Regulated MAP Kinases / metabolism
  • Flow Cytometry
  • Gene Expression Regulation / drug effects
  • Humans
  • Intestinal Mucosa / metabolism*
  • Liver / microbiology
  • Luminescent Agents / metabolism
  • Lung / microbiology
  • MAP Kinase Signaling System / drug effects
  • Mice
  • Mice, Inbred C57BL
  • Peritonitis / etiology
  • Peritonitis / metabolism
  • Peritonitis / microbiology*
  • Permeability
  • Phosphorylation

Substances

  • Luminescent Agents
  • Extracellular Signal-Regulated MAP Kinases
  • Alkaline Phosphatase

Associated data

  • Dryad/10.5061/dryad.13353

Grant support

Funding for this work came from the China Health and Medical Development Foundation (Grant: Research on Surgical Infection Mechanism and Control) and Research Fund of Peking University First Hospital, Grant No. 201196005.