Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Emma Millhouse is active.

Publication


Featured researches published by Emma Millhouse.


Fems Yeast Research | 2015

Polymicrobial Candida biofilms: friends and foe in the oral cavity

Lindsay E. O'Donnell; Emma Millhouse; Leighann Sherry; Ryan Kean; J. Malcolm; Christopher J. Nile; Gordon Ramage

The role of polymicrobial biofilm infections in medicine is becoming more apparent. Increasing number of microbiome studies and deep sequencing has enabled us to develop a greater understanding of how positive and negative microbial interactions influence disease outcomes. An environment where this is particularly pertinent is within the oral cavity, a rich and diverse ecosystem inhabited by both bacteria and yeasts, which collectively occupy and coexist within various niches as biofilm communities. Studies within this environment have however tended to be subject to extensive independent investigation, in the context of either polymicrobial bacterial communities or yeast biofilms, but rarely both together. It is clear however that they are not mutually exclusive. Therefore, this review aims to explore the influence of candidal populations on the composition of these complex aggregates and biofilm communities, to investigate their mechanistic interactions to understand how these impact clinical outcomes, and determine whether we can translate how this knowledge can be used to improve patient management.


BMC Oral Health | 2014

Development of an in vitro periodontal biofilm model for assessing antimicrobial and host modulatory effects of bioactive molecules

Emma Millhouse; Anto Jose; Leighann Sherry; David F. Lappin; Nisha Patel; Andrew M. Middleton; Jonathan Pratten; Shauna Culshaw; Gordon Ramage

BackgroundInflammation within the oral cavity occurs due to dysregulation between microbial biofilms and the host response. Understanding how different oral hygiene products influence inflammatory properties is important for the development of new products. Therefore, creation of a robust host-pathogen biofilm platform capable of evaluating novel oral healthcare compounds is an attractive option. We therefore devised a multi-species biofilm co-culture model to evaluate the naturally derived polyphenol resveratrol (RSV) and gold standard chlorhexidine (CHX) with respect to anti-biofilm and anti-inflammatory properties.MethodsAn in vitro multi-species biofilm containing S. mitis, F. nucleatum, P. gingivalis and A. actinomycetemcomitans was created to represent a disease-associated biofilm and the oral epithelial cell in OKF6-TERT2. Cytotoxicity studies were performed using RSV and CHX. Multi-species biofilms were either treated with either molecule, or alternatively epithelial cells were treated with these prior to biofilm co-culture. Biofilm composition was evaluated and inflammatory responses quantified at a transcriptional and protein level.ResultsCHX was toxic to epithelial cells and multi-species biofilms at concentrations ranging from 0.01-0.2%. RSV did not effect multi-species biofilm composition, but was toxic to epithelial cells at concentrations greater than 0.01%. In co-culture, CHX-treated biofilms resulted in down regulation of the inflammatory chemokine IL-8 at both mRNA and protein level. RSV-treated epithelial cells in co-culture were down-regulated in the release of IL-8 protein, but not mRNA.ConclusionsCHX possesses potent bactericidal properties, which may impact downstream inflammatory mediators. RSV does not appear to have bactericidal properties against multi-species biofilms, however it did appear to supress epithelial cells from releasing inflammatory mediators. This study demonstrates the potential to understand the mechanisms by which different oral hygiene products may influence gingival inflammation, thereby validating the use of a biofilm co-culture model.


BMC Oral Health | 2013

Investigating the biological properties of carbohydrate derived fulvic acid (CHD-FA) as a potential novel therapy for the management of oral biofilm infections

Leighann Sherry; Emma Millhouse; David F. Lappin; Colin Murray; Shauna Culshaw; Christopher J. Nile; Gordon Ramage

BackgroundA number of oral diseases, including periodontitis, derive from microbial biofilms and are associated with increased antimicrobial resistance. Despite the widespread use of mouthwashes being used as adjunctive measures to control these biofilms, their prolonged use is not recommended due to various side effects. Therefore, alternative broad-spectrum antimicrobials that minimise these effects are highly sought after. Carbohydrate derived fulvic acid (CHD-FA) is an organic acid which has previously demonstrated to be microbiocidal against Candida albicans biofilms, therefore, the aims of this study were to evaluate the antibacterial activity of CHD-FA against orally derived biofilms and to investigate adjunctive biological effects.MethodsMinimum inhibitory concentrations were evaluated for CHD-FA and chlorhexidine (CHX) against a range of oral bacteria using standardised microdilution testing for planktonic and sessile. Scanning electron microscopy was also employed to visualise changes in oral biofilms after antimicrobial treatment. Cytotoxicity of these compounds was assessed against oral epithelial cells, and the effect of CHD-FA on host inflammatory markers was assessed by measuring mRNA and protein expression.ResultsCHD-FA was highly active against all of the oral bacteria tested, including Porphyromonas gingivalis, with a sessile minimum inhibitory concentration of 0.5%. This concentration was shown to kill multi-species biofilms by approximately 90%, levels comparable to that of chlorhexidine (CHX). In a mammalian cell culture model, pretreatment of epithelial cells with buffered CHD-FA was shown to significantly down-regulate key inflammatory mediators, including interleukin-8 (IL-8), after stimulation with a multi-species biofilm.ConclusionsOverall, CHD-FA was shown to possess broad-spectrum antibacterial activity, with a supplementary function of being able to down-regulate inflammation. These properties offer an attractive spectrum of function from a naturally derived compound, which could be used as an alternative topical treatment strategy for oral biofilm diseases. Further studies in vitro and in vivo are required to determine the precise mechanism by which CHD-FA modulates the host immune response.


Journal of Periodontal Research | 2017

The epithelial cell response to health and disease associated oral biofilm models.

Gordon Ramage; David F. Lappin; Emma Millhouse; J. Malcolm; Anto Jose; J. Yang; David J. Bradshaw; Jonathan Pratten; Shauna Culshaw

Background and Objective Different bacteria differentially stimulate epithelial cells. Biofilm composition and viability are likely to influence the epithelial response. In vitro model systems are commonly used to investigate periodontitis‐associated bacteria and their interactions with the host; therefore, understanding factors that influence biofilm–cell interactions is essential. The present study aimed to develop in vitro monospecies and multispecies biofilms and investigate the epithelial response to these biofilms. Material and Methods Bacterial biofilms were cultured in vitro and then either live or methanol‐fixed biofilms were co‐cultured with epithelial cells. Changes in epithelial cell viability, gene expression and cytokine content of culture supernatants were evaluated. Results Bacterial viability was better preserved within mixed‐species biofilm culture than within single‐species biofilm culture. Both mixed‐ and single‐species biofilms stimulated increased expression of mRNA for interleukin 8 (IL8), C‐X‐C motif chemokine ligand 3 (CXCL3), C‐X‐C motif chemokine ligand 1 (CXCL1), interleukin 1 (IL1), interleukin 6 (IL6), colony‐stimulating factor 2 (CSF2) and tumour necrosis factor (TNF), and the response was greatest in response to mixed‐species biofilms. Following co‐culture, cytokines detected in the supernatants included IL‐8, IL‐6, granulocyte colony‐stimulating factor and granulocyte–macrophage colony‐stimulating factor, with the greatest release of cytokines found following co‐culture with methanol‐fixed, mixed‐species biofilms. Conclusions These data show that epithelial cells generate a distinct cytokine gene‐ and protein‐expression signature in response to live or fixed, single‐ or multispecies biofilms.


Journal of Dental Research | 2016

Mast Cells Contribute to Porphyromonas gingivalis–induced Bone Loss

J. Malcolm; Owain R. Millington; Emma Millhouse; L. Campbell; A. Adrados Planell; John Butcher; Catherine E. Lawrence; Kirsty Ross; Gordon Ramage; Iain B. McInnes; Shauna Culshaw

Periodontitis is a chronic inflammatory and bone-destructive disease. Development of periodontitis is associated with dysbiosis of the microbial community, which may be caused by periodontal bacteria, such as Porphyromonas gingivalis. Mast cells are sentinels at mucosal surfaces and are a potent source of inflammatory mediators, including tumor necrosis factors (TNF), although their role in the pathogenesis of periodontitis remains to be elucidated. This study sought to determine the contribution of mast cells to local bone destruction following oral infection with P. gingivalis. Mast cell–deficient mice (KitW-sh/W-sh) were protected from P. gingivalis–induced alveolar bone loss, with a reduction in anti–P. gingivalis serum antibody titers compared with wild-type infected controls. Furthermore, mast cell–deficient mice had reduced expression of Tnf, Il6, and Il1b mRNA in gingival tissues compared with wild-type mice. Mast cell–engrafted KitW-sh/W-sh mice infected with P. gingivalis demonstrated alveolar bone loss and serum anti–P. gingivalis antibody titers equivalent to wild-type infected mice. The expression of Tnf mRNA in gingival tissues of KitW-sh/W-sh mice was elevated following the engraftment of mast cells, indicating that mast cells contributed to the Tnf transcript in gingival tissues. In vitro, mast cells degranulated and released significant TNF in response to oral bacteria, and neutralizing TNF in vivo abrogated alveolar bone loss following P. gingivalis infection. These data indicate that mast cells and TNF contribute to the immunopathogenesis of periodontitis and may offer therapeutic targets.


Frontiers in Microbiology | 2016

Viable compositional analysis of an eleven species oral polymicrobial biofilm

Leighann Sherry; Gillian Lappin; Lindsay E. O'Donnell; Emma Millhouse; Owain R. Millington; David J. Bradshaw; Alyson S. Axe; Craig Williams; Christopher J. Nile; Gordon Ramage

Purpose: Polymicrobial biofilms are abundant in clinical disease, particularly within the oral cavity. Creating complex biofilm models that recapitulate the polymicrobiality of oral disease are important in the development of new chemotherapeutic agents. In order to do this accurately we require the ability to undertake compositional analysis, in addition to determine individual cell viability, which is difficult using conventional microbiology. The aim of this study was to develop a defined multispecies denture biofilm model in vitro, and to assess viable compositional analysis following defined oral hygiene regimens. Methods: An in vitro multispecies denture biofilm containing various oral commensal and pathogenic bacteria and yeast was created on poly (methyl methacrylate) (PMMA). Denture hygiene regimens tested against the biofilm model included brushing only, denture cleansing only and combinational brushing and denture cleansing. Biofilm composition and viability were assessed by culture (CFU) and molecular (qPCR) methodologies. Scanning electron microscopy and confocal laser scanning microscopy were also employed to visualize changes in denture biofilms following treatment. Results: Combinational treatment of brushing and denture cleansing had the greatest impact on multispecies denture biofilms, reducing the number of live cells by more than 2 logs, and altering the overall composition in favor of streptococci. This was even more evident during the sequential testing, whereby daily sequential treatment reduced the total and live number of bacteria and yeast more than those treated intermittently. Bacteria and yeast remaining following treatment tended to aggregate in the pores of the PMMA, proving more difficult to fully eradicate the biofilm. Conclusions: Overall, we are the first to develop a method to enable viable compositional analysis of an 11 species denture biofilm following chemotherapeutic challenge. We were able to demonstrate viable cell reduction and changes in population dynamics following evaluation of various denture cleansing regimens. Specifically, it was demonstrated that daily combinational treatment of brushing and cleansing proved to be the most advantageous denture hygiene regimen, however, residual organisms still remained within the pores of PMMA surface, which could act as a reservoir for further biofilm regrowth. We have identified an industry need for denture cleansing agents with the capacity to penetrate these pores and disaggregate these complex biofilm consortia.


Molecular Oral Microbiology | 2016

T cells, teeth and tissue destruction - what do T cells do in periodontal disease?

Lauren Campbell; Emma Millhouse; J. Malcolm; Shauna Culshaw

The microbial plaque biofilm resides adjacent to the tissue-destructive inflammatory infiltrate in periodontitis. Although not sufficient, this biofilm is necessary for this inflammatory response. Patients with periodontitis generate antibodies specific for bacteria in the biofilm - although the role of these antibodies is not clear, there is, undoubtedly, an adaptive immune response in periodontitis. T lymphocytes are central to adaptive immunity, and provide help for B cells to generate specific antibodies. T-cell receptor recognition of peptide antigen in the context of major histocompatibility complex can result in T-cell activation. The activation and differentiation of the T-cell can take many forms, and hence numerous types of T cells have been described. The role of adaptive immune responses, and the T-cell component thereof, in periodontitis remains relatively poorly defined. This review aims to broadly summarize findings about T cells and their role in periodontitis, focusing primarily on studies of human disease with a short discussion of some animal studies.


Molecular Oral Microbiology | 2017

The Porphyromonas gingivalis hemagglutinins HagB and HagC are major mediators of adhesion and biofilm formation.

Eimear Connolly; Emma Millhouse; Rebecca Doyle; Shauna Culshaw; Gordon Ramage; Gary P. Moran

Porphyromonas gingivalis is a bacterium associated with chronic periodontitis that possesses a family of genes encoding hemagglutinins required for heme acquisition. In this study we generated ΔhagB and ΔhagC mutants in strain W83 and demonstrate that both hagB and hagC are required for adherence to oral epithelial cells. Unexpectedly, a double ΔhagB/ΔhagC mutant had less severe adherence defects than either of the single mutants, but was found to exhibit increased expression of the gingipain-encoding genes rgpA and kgp, suggesting that a ΔhagB/ΔhagC mutant is only viable in populations of cells that exhibit increased expression of genes involved in heme acquisition. Disruption of hagB in the fimbriated strain ATCC33277 demonstrated that HagB is also required for stable attachment of fimbriated bacteria to oral epithelial cells. Mutants of hagC were also found to form defective single and multi-species biofilms that had reduced biomass relative to biofilms formed by the wild-type strain. This study highlights the hitherto unappreciated importance of these genes in oral colonization and biofilm formation.


PLOS ONE | 2016

In Vitro Effect of Porphyromonas gingivalis Methionine Gamma Lyase on Biofilm Composition and Oral Inflammatory Response.

Abish S. Stephen; Emma Millhouse; Leighann Sherry; Joseph Aduse-Opoku; Shauna Culshaw; Gordon Ramage; David J. Bradshaw; Gary R. Burnett; Robert P. Allaker

Methanethiol (methyl mercaptan) is an important contributor to oral malodour and periodontal tissue destruction. Porphyromonas gingivalis, Prevotella intermedia and Fusobacterium nucleatum are key oral microbial species that produce methanethiol via methionine gamma lyase (mgl) activity. The aim of this study was to compare an mgl knockout strain of P. gingivalis with its wild type using a 10-species biofilm co-culture model with oral keratinocytes and its effect on biofilm composition and inflammatory cytokine production. A P. gingivalis mgl knockout strain was constructed using insertion mutagenesis from wild type W50 with gas chromatographic head space analysis confirming lack of methanethiol production. 10-species biofilms consisting of Streptococcus mitis, Streptococcus oralis, Streptococcus intermedius, Fusobacterium nucleatum ssp polymorphum, Fusobacterium nucleatum ssp vincentii, Veillonella dispar, Actinomyces naeslundii, Prevotella intermedia and Aggregatibacter actinomycetemcomitans with either the wild type or mutant P. gingivalis were grown on Thermanox cover slips and used to stimulate oral keratinocytes (OKF6-TERT2), under anaerobic conditions for 4 and 24 hours. Biofilms were analysed by quantitative PCR with SYBR Green for changes in microbial ecology. Keratinocyte culture supernatants were analysed using a multiplex bead immunoassay for cytokines. Significant population differences were observed between mutant and wild type biofilms; V. dispar proportions increased (p<0.001), whilst A. naeslundii (p<0.01) and Streptococcus spp. (p<0.05) decreased in mutant biofilms. Keratinocytes produced less IL-8, IL-6 and IL-1α when stimulated with the mutant biofilms compared to wild type. Lack of mgl in P. gingivalis has been shown to affect microbial ecology in vitro, giving rise to a markedly different biofilm composition, with a more pro-inflammatory cytokine response from the keratinocytes observed. A possible role for methanethiol in biofilm formation and cytokine response with subsequent effects on oral malodor and periodontitis is suggested.


Frontiers in Microbiology | 2018

Evaluating Streptococcus mutans Strain Dependent Characteristics in a Polymicrobial Biofilm Community

Yan Zhou; Emma Millhouse; Tracy Shaw; David F. Lappin; Ranjith Rajendran; Jeremy Bagg; Huancai Lin; Gordon Ramage

Aim: The purpose of this study was to investigate strain dependent differences of the cariogenic biofilm forming Streptococcus mutans within both simple and complex communities. Methods: A mono-species containing representative S. mutans clinical isolates (caries and non-caries), and a multispecies in vitro caries biofilm model containing Lactobacillus casei, Veillonella dispar, Fusobacterium nucleatum and Actinomyces naeslundii, and either of two representative S. mutans clinical isolates (caries and non-caries), was developed as a comparison model. Compositional analysis of total and live bacteria within biofilms, and transcriptional analysis of biofilm associated virulence factors were evaluated by live/dead PCR and quantitative PCR, respectively. Scanning electron microscopy (SEM) was used to analyze the architecture of biofilm. One-way analysis of variance and t-tests were used to investigate significant differences between independent groups of data. Results: Within a mono-species biofilm, different S. mutans strains responded similarly to one another during biofilm formation in different carbohydrate sources, with sucrose showing the highest levels of biofilm biomass and galactose showing the lowest. Within the polymicrobial biofilm system, compositional analysis of the bacteria within the biofilm showed that S. mutans derived from a caries-free patient was preferentially composed of both total and viable L. casei, whereas S. mutans derived from a caries patient was dominated by both total and viable S. mutans (p < 0.001). Normalized gene expression analysis of srtA, gtfB, ftf, spaP, gbpB, and luxS, showed a general upregulation within the S. mutans dominant biofilm. Conclusion: We were able to demonstrate that individual strains derived from different patients exhibited altered biofilm characteristics, which were not obvious within a simple mono-species biofilm model. Influencing the environmental conditions changed the composition and functionality S. mutans within the polymicrobial biofilm. The biofilm model described herein provides a novel and reproducible method of assessing the impact on the biofilm microbiome upon different environmental influences.

Collaboration


Dive into the Emma Millhouse's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anto Jose

University of Glasgow

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge