Abstract

Macrophages play a key role in orchestrating the host immune response toward invading organisms or non-self molecules in the oral mucosa. Three-dimensional (3D) oral mucosal equivalents (OME) containing oral fibroblasts and keratinocytes are used extensively to mimic the human oral mucosa where they have been employed to examine innate immune responses to both bacterial and fungal pathogens as well as to biomaterials. Although the presence of immune cells is critical in generating an immune response, very few studies have incorporated leukocytes into OME, and to date, none have contained primary human macrophages. In this study, we report the generation of an immunocompetent OME to investigate immune responses toward bacterial challenge. Primary human monocyte-derived macrophages (MDM) were as responsive to bacterial lipopolysaccharide (LPS) challenge when cultured within a 3D hydrogel in terms of proinflammatory cytokine (IL-6, CXCL8, and TNF-α) gene expression and protein secretion compared with culture as two-dimensional monolayers. MDM were incorporated into a type 1 collagen hydrogel along with oral fibroblasts and the apical surface seeded with oral keratinocytes to generate an MDM-containing OME. Full-thickness MDM-OME displayed a stratified squamous epithelium and a fibroblast-populated connective tissue containing CD68-positive MDM that could be readily isolated to a single-cell population for further analysis by collagenase treatment followed by flow cytometry. When stimulated with LPS, MDM-OME responded with increased proinflammatory cytokine secretion, most notably for TNF-α that increased 12-fold when compared with OME alone. Moreover, this proinflammatory response was inhibited by pretreatment with dexamethasone, showing that MDM-OME are also amenable to drug treatment. Dual-labeled immunofluorescence confocal microscopy revealed that MDM were the sole source of TNF-α production within MDM-OME. These data show functional activity of MDM-OME and illustrate their usefulness for investigations aimed at monitoring the immune response of the oral mucosa to pathogens, biomaterials, and for tissue toxicity and anti-inflammatory drug delivery studies.Impact statementThree-dimensional in vitro models of the oral mucosa have been used extensively to investigate the host response to pathogens, but, to date, few have contained primary leukocytes. In this report, we describe the successful incorporation of primary human macrophages into oral mucosal equivalents (OME). These macrophage-containing models were histologically similar to the oral mucosa and immunoresponsive to bacterial lipopolysaccharides by upregulation of key proinflammatory markers. These advanced OME will significantly aid research into host–pathogen interaction, biomaterial toxicity, and drug delivery studies where the presence of an immune cell component is critical to better represent host oral tissue.

Highlights

  • Tissue-engineered oral mucosal equivalents (OME) have been used extensively to study the oral mucosa as improved model systems compared with in vitro cultured oral keratinocytes grown as two-dimensional (2D) monolayers.[1]

  • Resident and recruited immune cells are essential for maintenance of oral tissue and are critical in driving host responses to external insults, while dysregulation of the immune response can cause chronic conditions leading to debilitating oral lesions or poor outcomes in the case of oral squamous cell carcinoma.[13,14,15]

  • Normal oral fibroblasts (NOF) were isolated from the connective tissue of biopsies obtained from the oral mucosa of patients during routine dental procedures with written informed consent (Ethical Approval No 09/H1308/66) and cultured in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% v/v fetal bovine serum (FBS), 2 mM L-glutamine, 100 IU/mL penicillin, and 100 mg/mL streptomycin.[6]

Read more

Summary

Introduction

Tissue-engineered oral mucosal equivalents (OME) have been used extensively to study the oral mucosa as improved model systems compared with in vitro cultured oral keratinocytes grown as two-dimensional (2D) monolayers.[1]. Macrophage activation can occur through recognition of pathogen-associated molecular patterns, for example, in response to lipopolysaccharides (LPS) of gram-negative bacteria by cell surface pattern recognition receptors, such as CD14 and Toll-like receptors This interaction induces intracellular signaling leading to increased gene expression and secretion of proinflammatory cytokines such as TNF-a16 that can be inhibited by anti-inflammatory therapeutics such as glucocorticoids.[17] Addition of immune cells, such as macrophages, to current OME would increase their sensitivity to detect and respond to foreign molecules, making them more representative of the native oral mucosa. We describe the generation of a tissue-engineered immune model of nonkeratinized oral mucosa containing primary human monocyte-derived macrophages (MDM) These immune OME produced functional responses upon stimulation with Escherichia coli LPS in the form of increased secretion of proinflammatory cytokines that were inhibited by dexamethasone treatment. These OME will be extremely beneficial for studies aimed at investigating macrophage behavior in disease and cellular toxicity, as well as for examining immune response to drug and/or biomaterials exposure

Methods
Findings
Discussion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call