Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Front Oral Health ; 2: 604565, 2021.
Article in English | MEDLINE | ID: mdl-35047989

ABSTRACT

The incidence of human papillomavirus (HPV)-associated cancer is increasing and HPV is now implicated in the aetiology of more than 60% of all oropharyngeal squamous cell carcinomas (OPSCC). In OPSCC, innate immune cells such as neutrophils and macrophages generally correlate with poor prognosis, whilst adaptive immune cells, such as lymphocytes, tend to correlate with improved prognosis. This may, in part, be due to differences in the immune response within the tumour microenvironment leading to the recruitment of specific tumour-associated leukocyte sub-populations. In this study, we aimed to examine if differences exist in the levels of infiltrated leukocyte sub-populations, with particular emphasis on tumour-associated neutrophils (TAN), and to determine the mechanism of chemokine-induced leukocyte recruitment in HPV-positive compared to HPV-negative OPSCC. Immunohistochemical analysis showed that HPV-negative OPSCC contained significantly more neutrophils than HPV-positive tumours, whilst levels of CD68+ macrophages and CD3+ lymphocytes were similar. Using a 3D tissue culture model to represent tumour-stromal interactions, we demonstrated that HPV-negative tumour-stromal co-cultures expressed significantly higher levels of CXCL8, leading to increased neutrophil recruitment compared to their HPV-positive counterparts. HPV-negative OPSCC cells have previously been shown to express higher levels of IL-1 than their HPV-positive counterparts, indicating that this cytokine may be responsible for driving increased chemokine production in the HPV-negative 3D model. Inhibition of IL-1R in the tumour-stromal models using the receptor-specific antagonist, anakinra, dramatically reduced chemokine secretion and significantly impaired neutrophil and monocyte recruitment, suggesting that this tumour-stromal response is mediated by the IL-1/IL-1R axis. Here, we identify a mechanism by which HPV-negative OPSCC may recruit more TAN than HPV-positive OPSCC. Since TAN are associated with poor prognosis in OPSCC, our study identifies potential therapeutic targets aimed at redressing the chemokine imbalance to reduce innate immune cell infiltration with the aim of improving patient outcome.

2.
Int J Cancer ; 144(2): 334-344, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30191960

ABSTRACT

Human papillomavirus (HPV) is now recognised as a major aetiological agent in the pathogenesis of oropharyngeal carcinoma (OPC). HPV-positive tumours are associated with better outcomes compared to HPV-negative tumours, possibly due to differences in their aetiology and/or the tumour microenvironment. Increased numbers of tumour-associated leukocytes have been observed in many cancers including OPC, with variable influence on prognosis depending on the leukocyte subpopulation investigated. Whether HPV status influences leukocyte recruitment to OPC remains unknown. This in-vitro study examined differences in the chemoattractant capacity of HPV-positive and HPV-negative OPC cell lines. Gene and protein expression analysis demonstrated that whilst both monocultures of HPV-positive and HPV-negative cell lines, along with normal tonsillar fibroblasts (NTF), expressed low chemokine levels, NTF cultured with conditioned medium from HPV-negative OPC cells expressed significantly higher levels of all chemokines tested compared to NTF incubated with the medium from HPV-positive OPC cell lines. HPV-negative OPC lines expressed IL-1ß mRNA whereas HPV-positive cells did not, and NTF constitutively expressed IL-1R1. Pre-treatment with the IL-R antagonist, anakinra or siRNA to IL-1R1 significantly reduced chemokine secretion from NTF stimulated with conditioned medium from HPV-negative tumour cells or recombinant IL-1ß (p < 0.05). These data suggest that secretion of chemokines is driven by the interaction between HPV-negative OPC cells and stromal fibroblasts through an IL-1/IL-1R-mediated mechanism that is less prominent within the HPV-positive tumour microenvironment. These observations may explain differences in leukocyte sub-populations recruited to HPV-positive versus negative OPC and indicate that HPV status is a key determinant in controlling the inflammatory tumour microenvironment.


Subject(s)
Fibroblasts/metabolism , Interleukin-1/metabolism , Oropharyngeal Neoplasms/pathology , Oropharyngeal Neoplasms/virology , Receptors, Interleukin-1 Type I/metabolism , Cell Line, Tumor , Chemokines/metabolism , Chemotaxis, Leukocyte/physiology , Humans , Oropharyngeal Neoplasms/metabolism , Papillomavirus Infections , Tumor Microenvironment/physiology
3.
Tissue Cell ; 52: 71-77, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29857831

ABSTRACT

Recent advances in three-dimensional printing technology have led to a rapid expansion of its applications in tissue engineering. The present study was designed to develop and characterize an in vitro multi-layered human alveolar bone, based on a 3D printed scaffold, combined with tissue engineered oral mucosal model. The objective was to incorporate oral squamous cell carcinoma (OSCC) cell line spheroids to the 3D model at different anatomical levels to represent different stages of oral cancer. Histological evaluation of the 3D tissue model revealed a tri-layered structure consisting of distinct epithelial, connective tissue, and bone layers; replicating normal oral tissue architecture. The mucosal part showed a well-differentiated stratified oral squamous epithelium similar to that of the native tissue counterpart, as demonstrated by immunohistochemistry for cytokeratin 13 and 14. Histological assessment of the cancerous models demonstrated OSCC spheroids at three depths including supra-epithelial level, sub-epithelial level, and deep in the connective tissue-bone interface. The 3D tissue engineered composite model closely simulated the native oral hard and soft tissues and has the potential to be used as a valuable in vitro model for the investigation of bone invasion of oral cancer and for the evaluation of novel diagnostic or therapeutic approaches to manage OSCC in the future.


Subject(s)
Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/pathology , Models, Anatomic , Mouth Neoplasms/pathology , Printing, Three-Dimensional , Tissue Engineering/methods , Alveolar Process/pathology , Humans , Spheroids, Cellular , Squamous Cell Carcinoma of Head and Neck , Tissue Scaffolds , Tumor Cells, Cultured
4.
Tissue Eng Part C Methods ; 24(2): 99-107, 2018 02.
Article in English | MEDLINE | ID: mdl-29092692

ABSTRACT

Advances in tissue engineering have permitted assembly of multilayered composite tissue constructs for potential applications in the treatment of combined hard and soft tissue defects and as an alternative in vitro test model to animal experimental systems. The aim of this study was to develop and characterize a novel three-dimensional combined human alveolar bone and gingival mucosal model based on primary cells isolated from the oral tissues. Bone component of the model was engineered by seeding primary human alveolar osteoblasts into a hydroxyapatite/tricalcium phosphate scaffold and culturing in a spinner bioreactor. The engineered bone was then laminated, using an adhesive tissue sealant, with tissue-engineered gingival mucosa consisting of air/liquid interface-cultured normal human gingival keratinocytes on oral fibroblast-populated collagen gel scaffold. Histological characterization revealed a structure consisting of established epithelial, connective tissue and bone layers closely comparable to normal oral tissue architecture. The mucosal component demonstrated a mature epithelium undergoing terminal differentiation similar to that characteristic of native buccal mucosa, as confirmed using cytokeratin 13 and cytokeratin 14 immunohistochemistry. Ultrastructural analysis confirmed the presence of desmosomes and hemidesmosomes in the epithelial layer, a continuous basement membrane, and newly synthesized collagen in the connective tissue layer. Quantitative polymerase chain reaction (qPCR) assessment of osteogenesis-related gene expression showed a higher expression of genes encoded collagen I (COL1) and osteonectin (ON) compared with osteocalcin (OC), osteopontin (OP), and alkaline phosphatase (ALP). Enzyme-linked immunosorbent assay quantification of COL1, ON, and OC confirmed a pattern of secretion, which paralleled the model's gene expression profile. We demonstrate in this study that, replicating the anatomical setting between oral mucosa and the underlying alveolar bone is feasible and the developed model showed characteristics similar to those of normal tissue counterparts. This trilayered model therefore offers great scope as an advanced and anatomically representative tissue-engineered alternative to animal models.


Subject(s)
Alveolar Process/cytology , Bone Regeneration , Fibroblasts/cytology , Gingiva/cytology , Mouth Mucosa/cytology , Osteoblasts/cytology , Tissue Engineering/methods , Alveolar Process/metabolism , Biomarkers/metabolism , Cells, Cultured , Fibroblasts/metabolism , Gingiva/metabolism , Humans , Mouth Mucosa/metabolism , Osteoblasts/metabolism , Tissue Scaffolds
SELECTION OF CITATIONS
SEARCH DETAIL
...