Pomegranate Extract Affects Gut Biofilm Forming Bacteria and Promotes Intestinal Mucosal Healing Regulating the Crosstalk between Epithelial Cells and Intestinal Fibroblasts

Nutrients. 2023 Apr 5;15(7):1771. doi: 10.3390/nu15071771.

Abstract

Background: Pomegranate (Punica granatum) can be used to prepare a bioactive extract exerting anti-inflammatory activities. Clinical studies demonstrated an improvement in clinical response in inflammatory bowel disease (IBD) patients when pomegranate extract (PG) was taken as a complement to standard medications. However, the molecular mechanisms underlying its beneficial effects are still scarcely investigated. This study investigates the effect of PG on bacterial biofilm formation and the promotion of mucosal wound healing. Methods: The acute colitis model was induced in C57BL/6N mice by 3% dextran sodium sulfate administration in drinking water for 5 days. During the recovery phase of colitis, mice received saline or PG (200 mg/kg body weight) by oral gavage for 11 days. Colitis was scored daily by evaluating body weight loss, bleeding, and stool consistency. In vivo intestinal permeability was evaluated by fluorescein isothiocyanate-conjugated dextran assay, bacterial translocation was assessed by fluorescence in situ hybridization on tissues, whereas epithelial and mucus integrity were monitored by immunostaining for JAM-A and MUC-2 markers. Bacterial biofilm formation was assessed using microfluidic devices for 24 or 48 h. Primary fibroblasts were isolated from healthy and inflamed areas of 8 IBD patients, and Caco-2 cells were stimulated with or without PG (5 μg/mL). Inflammatory mediators were measured at the mRNA and protein level by RT-PCR, WB, or Bio-plex multiplex immunoassay, respectively. Results: In vivo, PG boosted the recovery phase of colitis, promoting a complete restoration of the intestinal barrier with the regeneration of the mucus layer, as also demonstrated by the absence of bacterial spread into the mucosa and the enrichment of crypt-associated fibroblasts. Microfluidic experiments did not highlight a specific effect of PG on Enterobacterales biofilm formation, even though Citrobacter freundii biofilm was slightly impaired in the presence of PG. In vitro, inflamed fibroblasts responded to PG by downregulating the release of metalloproteinases, IL-6, and IL-8 and upregulating the levels of HGF. Caco-2 cells cultured in a medium supplemented with PG increased the expression of SOX-9 and CD44, whereas in the presence of HGF or plated with a fibroblast-conditioned medium, they displayed a decrease in SOX-9 and CD44 expression and an increase in AXIN2, a negative regulator of Wnt signaling. Conclusions: These data provide new insight into the manifold effects of PG on promoting mucosal homeostasis in IBD by affecting pathogen biofilm formation and favoring the regeneration of the intestinal barrier through the regulation of the crosstalk between epithelial and stromal cells.

Keywords: Crohn’s disease; biofilm; mucosal healing; polyphenols; ulcerative colitis.

MeSH terms

  • Animals
  • Bacteria / genetics
  • Caco-2 Cells
  • Colitis* / chemically induced
  • Colitis* / drug therapy
  • Colitis* / genetics
  • Dextran Sulfate / pharmacology
  • Dextrans / therapeutic use
  • Disease Models, Animal
  • Epithelial Cells / metabolism
  • Humans
  • In Situ Hybridization, Fluorescence
  • Inflammatory Bowel Diseases* / metabolism
  • Intestinal Mucosa / metabolism
  • Mice
  • Mice, Inbred C57BL
  • Pomegranate*
  • Wound Healing

Substances

  • fluorescein isothiocyanate dextran
  • Dextrans
  • Dextran Sulfate

Grants and funding

This research was supported by: the ECCO Fellowship to S.E.; the Fondazione Umberto Veronesi Fellowship, and the European Union’s Horizon 2020 research and innovator program under the Marie Sklodowska-Curie grant agreement #101029427 to S.L.; and Esprinet SpA Italy and Associazione Italiana Ricerca contro il Cancro (AIRC), MFAG 2015 Id.17795 to S.V.