Intravaginal poly-(D, L-lactic-co-glycolic acid)-(polyethylene glycol) drug-delivery nanoparticles induce pro-inflammatory responses with Candida albicans infection in a mouse model

PLoS One. 2020 Oct 22;15(10):e0240789. doi: 10.1371/journal.pone.0240789. eCollection 2020.

Abstract

In a recent study, using an in vitro model to study intravaginal nanoparticle exposure during yeast infections, we demonstrated that C. albicans exposure suppressed apoptotic gene expression and induced oxidative stress and pyroptosis in vaginal epithelial cells. The mucous-penetrating drug delivery nanoparticles made from poly-(D, L-lactic-co-glycolic acid)-(polyethylene glycol) induced cytotoxicity by activating apoptosis, endoplasmic reticulum (ER) stress, oxidative stress, and DNA damage repair responses alone and, in some cases with C. albicans. In the current study we evaluated the effects of fluorescently-labelled nanoparticles in CBA/J mice challenged intravaginally for two hours followed by intravaginal challenge with C. albicans for 18 hours. Nanoparticle treatment increased systemic translocation of C. albicans threefold in the heart. C. albicans also increased systemic distribution of the nanoparticles fivefold in the heart. Flow cytometric assays showed co-localization of the nanoparticles with epithelial cells, macrophages and dendritic cells. Nanoparticle-treated, C. albicans-infected mice exhibited induction of autophagy, ER stress, apoptosis, and inflammatory serum cytokines. C. albicans infection was associated with pyroptosis and suppressed expression of ER stress and apoptosis-related genes. Induction of apoptosis during nanoparticle treatment and in nanoparticle-treated-C. albicans infected mice was observed as DNA damage responses, mitochondrial depolarization and (Poly [ADP-Ribose] Polymerase) cleavage. C. albicans infection was associated with increased mRNA expression of anti-apoptotic genes. Both C. albicans infection and nanoparticle treatment showed enhanced chemoattraction of dendritic cells and polymorphonuclear cells to factors in vaginal washings in a chemotaxis assay. This study shows that both intravaginal treatment of mice with the nanoparticles and infection with C. albicans induce cytotoxic and inflammatory responses. C. albicans also suppressed cell apoptosis. These results clarify our understanding of how nanoparticles modulate host cellular responses during C. albicans infection and will be applicable for future research and development of intravaginal nanomedicines.

Publication types

  • Research Support, U.S. Gov't, Non-P.H.S.
  • Research Support, U.S. Gov't, P.H.S.

MeSH terms

  • Animals
  • Apoptosis
  • Candida albicans / physiology*
  • Candidiasis / drug therapy*
  • Candidiasis / genetics
  • Candidiasis / microbiology
  • Cell Line
  • Chemotaxis
  • Cytokines / metabolism
  • Dendritic Cells / pathology
  • Disease Models, Animal
  • Drug Delivery Systems*
  • Epithelial Cells / microbiology
  • Epithelial Cells / pathology
  • Female
  • Gene Expression Regulation
  • Humans
  • Inflammation / genetics*
  • Inflammation Mediators / metabolism
  • Mice, Inbred CBA
  • Nanoparticles / chemistry*
  • Neutrophils / pathology
  • Organ Specificity
  • Polyethylene Glycols / chemistry*
  • Polylactic Acid-Polyglycolic Acid Copolymer / chemistry*
  • Stress, Physiological
  • Vagina / microbiology*
  • Vagina / pathology
  • Yeasts

Substances

  • Cytokines
  • Inflammation Mediators
  • Polylactic Acid-Polyglycolic Acid Copolymer
  • Polyethylene Glycols

Grants and funding

RDW received funding from the FDA Office of Women’s Health. TTL was supported in part by an appointment to the ORISE Research Participation Program at the National Center for Toxicological Research, U.S. Food and Drug Administration, administered by the Oak Ridge Institute for Science and Education through an interagency agreement between the U.S. Department of Energy and FDA/NCTR. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. There was no additional external funding received for this study.