Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Stem Cell Res Ther ; 15(1): 166, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38867276

ABSTRACT

BACKGROUND: Hypertrophic scarring results from myofibroblast differentiation and persistence during wound healing. Currently no effective treatment for hypertrophic scarring exists however, autologous fat grafting has been shown to improve scar elasticity, appearance, and function. The aim of this study was to understand how paracrine factors from adipose tissues and adipose-derived stromal cells (ADSC) affect fibroblast to myofibroblast differentiation. METHODS: The transforming growth factor-ß1 (TGF-ß1) induced model of myofibroblast differentiation was used to test the effect of conditioned media from adipose tissue, ADSC or lipid on the proportion of fibroblasts and myofibroblasts. RESULTS: Adipose tissue conditioned media inhibited the differentiation of fibroblasts to myofibroblasts but this inhibition was not observed following treatment with ADSC or lipid conditioned media. Hepatocyte growth factor (HGF) was readily detected in the conditioned medium from adipose tissue but not ADSC. Cells treated with HGF, or fortinib to block HGF, demonstrated that HGF was not responsible for the inhibition of myofibroblast differentiation. Conditioned media from adipose tissue was shown to reduce the proportion of myofibroblasts when added to fibroblasts previously treated with TGF-ß1, however, conditioned media treatment was unable to significantly reduce the proportion of myofibroblasts in cell populations isolated from scar tissue. CONCLUSIONS: Cultured ADSC or adipocytes have been the focus of most studies, however, this work highlights the importance of considering whole adipose tissue to further our understanding of fat grafting. This study supports the use of autologous fat grafts for scar treatment and highlights the need for further investigation to determine the mechanism.


Subject(s)
Adipose Tissue , Cell Differentiation , Hepatocyte Growth Factor , Myofibroblasts , Transforming Growth Factor beta1 , Myofibroblasts/metabolism , Myofibroblasts/drug effects , Myofibroblasts/cytology , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factor beta1/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Cell Differentiation/drug effects , Culture Media, Conditioned/pharmacology , Humans , Hepatocyte Growth Factor/pharmacology , Hepatocyte Growth Factor/metabolism , Paracrine Communication/drug effects , Phenotype , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/drug effects , Fibroblasts/cytology , Adipocytes/metabolism , Adipocytes/cytology , Adipocytes/drug effects , Stromal Cells/metabolism , Stromal Cells/cytology , Stromal Cells/drug effects
2.
J Physiol ; 601(22): 4895-4905, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37795936

ABSTRACT

Extracellular vesicles (EVs) are small, membrane-enclosed vesicles released by cells into the extracellular milieu. They are found in all body fluids and contain a variety of functional cargo including DNA, RNA, proteins, glycoproteins and lipids, able to provoke phenotypic responses in cells, both locally and at distant sites. They are implicated in a wide array of physiological and pathological processes and hence have attracted considerable attention in recent years as potential therapeutic targets, drug delivery vehicles and biomarkers of disease. In this review we summarise the major functions of EVs in health and disease and discuss their translational potential, highlighting opportunities of - and challenges to - capitalising on our rapidly increasing understanding of EV biology for patient benefit.


Subject(s)
Extracellular Vesicles , Humans , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Cell Communication/physiology , Biomarkers/metabolism , Proteins
3.
Anaerobe ; 80: 102696, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36642290

ABSTRACT

OBJECTIVE: The study aimed to investigate the role of the PGN2012 gene of the periodontitis contributing pathobiont Porphyromonas gingivalis. PGN2012 is a homolgue of TolC and is a gene our group previously showed was overexpressed in hyperinvasive cells. METHODS: The study used a combination of bioinformatics, knockout mutagenesis, growth experiments, biofilm assays and human cell invation assays to investigate PGN2012 function. RESULTS: Bioinformatics identified that PGN2012 is part of one of four TolC containing gene loci in P. gingivalis that we predicted may encode a metal resistance RND family tripartite pump, similar to those present in other Gram-negative bacteria, but which are not well understood in anaerobic bacteria. A ΔPGN2012 deletion displayed slightly reduced growth in liquid culture but did not effect biofilm formation or human cell invasion. When metal ions were included in the medium the mutant displayed significantly increased sensitivity to the divalent metal ions Zn2+ (500 µM), Co2+ (2 mM), and Cd2+(0.1 mM) but not Cu2+. CONCLUSIONS: We propose to rename the PGN2012-2014 genes czcCBA, which we suggest plays a role in intracellular stress resistance where zinc is often employed by host cells in antibacterial defence with implications for chronic infection in humans.


Subject(s)
Periodontitis , Porphyromonas gingivalis , Humans , Porphyromonas gingivalis/genetics , Periodontitis/microbiology , Anti-Bacterial Agents , Zinc , Operon
4.
Br J Cancer ; 117(6): 867-875, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28742795

ABSTRACT

BACKGROUND: The molecular mechanisms involved in the invasion of bone by oral squamous cell carcinomas (OSCC) are poorly understood, and little is known about the role of cancer-associated fibroblasts (CAF), the presence of which confers a poor prognosis. METHODS: Clinicopathological data from 277 OSCC cases involving bone resections were reviewed, and 32 cases thoroughly analysed histologically. Immunohistochemistry was used to examine αSMA, RANKL and OPG. Western blotting and qPCR were used to assess myofibroblast (CAF-like) differentiation, RANKL and OPG expression in vitro, and RANKL secretion was analysed by ELISA. Osteoclastogenesis was examined using TRAP staining, multinucleation and pit forming assays. RESULTS: Fibrous stroma intervened between tumour and bone in the majority of cases, with no direct contact between cancer cells and bone. RANKL and OPG, two proteins key to regulating bone resorption, were expressed in tumour cells as well as fibrous stroma adjacent to bone and αSMA-positive myofibroblastic CAF were consistently seen infiltrating into bone ahead of tumour cells. Human primary osteoblasts cultured with conditioned media from human OSCC-derived cells and human primary CAF showed a significant increase in RANKL and a decline in OPG mRNA expression. RANKL secretion was significantly increased in primary oral fibroblasts induced to differentiate into a CAF-like phenotype by transforming growth factor-ß1 (TGF-ß1) treatment and in primary CAF. Indirect co-culture of murine macrophages with conditioned media from CAF (experimentally derived and isolated from OSCCs) resulted in a marked increase in osteoclastogenesis (in excess of that provoked by cancer cells) determined by tartrate-resistant acid phosphatase activity, multinucleation and resorption pit formation. CONCLUSIONS: This study is the first to describe a functional role for CAFs in bone invasion and turnover, identifying a novel potential therapeutic target and diagnostic indicator in this difficult to treat bone invasive malignancy.


Subject(s)
Bone Neoplasms/pathology , Cancer-Associated Fibroblasts/physiology , Carcinoma, Squamous Cell/pathology , Mouth Neoplasms/pathology , Neoplasm Proteins/analysis , Actins/analysis , Bone Neoplasms/chemistry , Bone and Bones/chemistry , Bone and Bones/drug effects , Bone and Bones/pathology , Cancer-Associated Fibroblasts/chemistry , Carcinoma, Squamous Cell/chemistry , Cell Differentiation , Cell Line, Tumor , Humans , Mitochondrial Proteins/analysis , Mouth Neoplasms/chemistry , Neoplasm Invasiveness , Osteogenesis , Osteoprotegerin/analysis , RANK Ligand , RNA, Messenger/metabolism , Receptor Activator of Nuclear Factor-kappa B/analysis , Ribosomal Proteins/analysis , Transforming Growth Factor beta1/pharmacology
5.
Eur J Oral Sci ; 125(4): 247-257, 2017 08.
Article in English | MEDLINE | ID: mdl-28653423

ABSTRACT

Angiotensin II (Ang II) is the product of the proteolytic action of angiotensin-converting enzyme (ACE) on the precursor peptide, angiotensin I (Ang I). In addition to its vasoactive properties, Ang II is able to stimulate angiogenesis and act as a mitogen, promoting cellular proliferation. Recently, evidence has emerged that Ang II is also able to promote tumour invasion, a key step in the metastatic cascade, although the mechanisms by which it does so remain largely obscure. Here we show that Ang II is able to promote the invasion and migration of head and neck squamous cell carcinoma (HNSCC) cells both in an autocrine manner and by triggering stromal tumour-paracrine interactions. The effects of Ang II on autocrine and paracrine signalling pathways are mediated by angiotensin receptor 1 (AT1 R) and inhibited by angiotensin 1-7 (Ang 1-7), a peptide produced from Ang II by the action of angiotensin-converting enzyme 2 (ACE2). These data are the first to demonstrate a role for the renin-angiotensin system in oral carcinogenesis and raise the possibility of utilizing AT1 R receptor antagonists and/or Ang 1-7 as novel therapeutic agents for HNSCC.


Subject(s)
Angiotensin II/metabolism , Angiotensin I/pharmacology , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Peptide Fragments/pharmacology , Cell Culture Techniques , Cell Movement , Disease Progression , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Immunoblotting , Polymerase Chain Reaction , Renin-Angiotensin System/physiology , Signal Transduction , Transfection , Tumor Cells, Cultured
6.
J Oral Pathol Med ; 46(2): 82-88, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27237745

ABSTRACT

There is now compelling evidence that the tumour stroma plays an important role in the pathogenesis of cancers of epithelial origin. The pre-eminent cell type of the stroma is carcinoma-associated fibroblasts. These cells demonstrate remarkable heterogeneity with activation and senescence being common stress responses. In this review, we summarise the part that these cells play in cancer, particularly oral cancer, and present evidence to show that activation and senescence reflect a unified programme of fibroblast differentiation. We report advances concerning the senescent fibroblast metabolome, mechanisms of gene regulation in these cells and ways in which epithelial cell adhesion is dysregulated by the fibroblast secretome. We suggest that the identification of fibroblast stress responses may be a valuable diagnostic tool in the determination of tumour behaviour and patient outcome. Further, the fact that stromal fibroblasts are a genetically stable diploid cell population suggests that they may be ideal therapeutic targets and early work in this context is encouraging.


Subject(s)
Fibroblasts/physiology , Mouth Neoplasms/pathology , Cellular Senescence , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Metabolome , Mouth Neoplasms/metabolism , Mouth Neoplasms/physiopathology
7.
Br J Cancer ; 111(4): 807-16, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25010866

ABSTRACT

BACKGROUND: HOX gene expression is altered in many cancers; previous microarray revealed changes in HOX gene expression in head and neck squamous cell carcinoma (HNSCC), particularly HOXD10. METHODS: HOXD10 expression was assessed by qPCR and immunoblotting in vitro and by immunohistochemistry (IHC) in tissues. Low-expressing cells were stably transfected with HOXD10 and the phenotype assessed with MTS, migration and adhesion assays and compared with the effects of siRNA knockdown in high-HOXD10-expressing cells. Novel HOXD10 targets were identified using expression microarrays, confirmed by reporter assay, and validated in tissues using IHC. RESULTS: HOXD10 expression was low in NOKs, high in most primary tumour cells, and low in lymph node metastasis cells, a pattern confirmed using IHC in tissues. Overexpression of HOXD10 decreased cell invasion but increased proliferation, adhesion and migration, with knockdown causing reciprocal effects. There was no consistent effect on apoptosis. Microarray analysis identified several putative HOXD10-responsive genes, including angiomotin (AMOT-p80) and miR-146a. These were confirmed as HOXD10 targets by reporter assay. Manipulation of AMOT-p80 expression resulted in phenotypic changes similar to those on manipulation of HOXD10 expression. CONCLUSIONS: HOXD10 expression varies by stage of disease and produces differential effects: high expression giving cancer cells a proliferative and migratory advantage, and low expression may support invasion/metastasis, in part, by modulating AMOT-p80 levels.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Homeodomain Proteins/physiology , Transcription Factors/physiology , Angiomotins , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Disease Progression , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/pathology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , MicroRNAs/genetics , Microfilament Proteins , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Squamous Cell Carcinoma of Head and Neck , Transcriptome
8.
Mol Oral Microbiol ; 28(5): 366-78, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23714361

ABSTRACT

Porphyromonas gingivalis and Tannerella forsythia are gram-negative pathogens strongly associated with periodontitis. Their abilities to interact, invade and persist within host cells are considered crucial to their pathogenicity, but the mechanisms by which they subvert host defences are not well understood. In this study, we set out to investigate whether P. gingivalis and T. forsythia directly target key signalling molecules that may modulate the host cell phenotype to favour invasion and persistence. Our data identify, for the first time, that P. gingivalis, but not T. forsythia, reduces levels of intracellular mammalian target of rapamycin (mTOR) in oral epithelial cells following invasion over a 4-h time course, via the action of gingipains. The ability of cytochalasin D to abrogate P. gingivalis-mediated mTOR degradation suggests that this effect is dependent upon cellular invasion. We also show that levels of several other proteins in the mTOR signalling pathway are modulated by gingipains, either directly or as a consequence of mTOR degradation including p-4E-BP1. Taken together, our data suggest that P. gingivalis manipulates the mTOR pathway, providing evidence for a potentially novel mechanism by which P. gingivalis mediates its effects on host cell responses to infection.


Subject(s)
Adhesins, Bacterial/pharmacology , Cysteine Endopeptidases/pharmacology , Periodontitis/microbiology , Porphyromonas gingivalis/metabolism , TOR Serine-Threonine Kinases/drug effects , Adaptor Proteins, Signal Transducing/drug effects , Adhesins, Bacterial/drug effects , Bacteroidaceae Infections/microbiology , Bacteroides/metabolism , Bacteroides Infections/microbiology , Carrier Proteins/drug effects , Cell Line , Cell Line, Tumor , Cysteine Endopeptidases/drug effects , Cytochalasin D/pharmacology , Epithelial Cells/microbiology , Gingipain Cysteine Endopeptidases , Humans , Keratinocytes/microbiology , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mouth Mucosa/microbiology , Multiprotein Complexes/drug effects , Nucleic Acid Synthesis Inhibitors/pharmacology , Oncogene Protein v-akt/drug effects , Porphyromonas gingivalis/drug effects , Rapamycin-Insensitive Companion of mTOR Protein , Regulatory-Associated Protein of mTOR , Signal Transduction/drug effects
9.
J Oral Pathol Med ; 42(4): 309-14, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23173553

ABSTRACT

BACKGROUND: Exposure to factors released from tobacco during chewing or smoking is recognized as a major risk factor for oral carcinogenesis and influences the phenotype of oral epithelial cells and fibroblasts within the underlying stroma. Micro(mi)RNA can regulate the expression of genes within cells, and previous studies show that tobacco products can alter the miRNA profiles in lung epithelial cells. However, the molecular alterations occurring in oral fibroblasts exposed to tobacco constituents remain to be elucidated. METHODS: Oral fibroblasts were exposed to cigarette smoke condensate (CSC) and miRNA expression compared to untreated controls using tiling low-density arrays (TLDA). Expression of miRNA-145 was confirmed by quantitative (q)RT-PCR. The effect of CSC on fibroblast cell viability, motility and matrix metalloproteinase (MMP)-2 expression was measured using MTS, a wound scratch assay and qRT-PCR, respectively. Oral cancer cell migration in response to culture supernatants from mock, control or pre-miR-145-transfected CSC-treated fibroblasts was analysed by chemotaxis assay. RESULTS: TLDA analysis identified widespread changes in the miRNA expression profile of fibroblasts exposed to CSC. Pri-, pre- and mature miRNA-145 were significantly down-regulated in response to CSC, and this was accompanied by up-regulated expression of MMP-2 and increased migration of fibroblasts compared to untreated controls. Re-expression of miR-145 abrogated the ability of fibroblasts to promote oral cancer cell chemotaxis in response to CSC. CONCLUSION: These findings suggest that tobacco constituents influence the expression of miRNA within oral fibroblasts promoting a phenotype that increases oral cancer migration and sheds new light on the mechanisms underlying oral cancer pathogenesis.


Subject(s)
Cell Transformation, Neoplastic/chemically induced , Fibroblasts/drug effects , MicroRNAs/analysis , Mouth Mucosa/drug effects , Nicotiana/adverse effects , Smoke/adverse effects , Tobacco Products/adverse effects , Carcinoma, Squamous Cell/pathology , Cell Culture Techniques , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Transformation, Neoplastic/pathology , Chemotaxis/drug effects , Culture Media, Conditioned , Epithelial Cells/drug effects , Gene Expression Profiling , Humans , Matrix Metalloproteinase 2/drug effects , Microarray Analysis , Mouth Mucosa/cytology , Phenotype , RNA, Small Nuclear , Real-Time Polymerase Chain Reaction , Stromal Cells/drug effects , Transfection
10.
Life Sci ; 91(13-14): 557-61, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22525370

ABSTRACT

AIMS: The aims of this study were to examine the role of endothelin-1 (ET-1), a pleiotropic peptide found at elevated levels in a number of malignancies and which has been shown to influence oral cancer cell behaviour via paracrine signalling pathways, on the phenotype of oral fibroblasts. MAIN METHODS: The effect of ET-1 on proliferation and migration of human primary oral fibroblasts was assessed using MTS and scratch assays, respectively. The ability of ET-1 to affect fibroblast contractility was analysed using type-I collagen gels. Changes in gene expression in oral fibroblasts exposed to ET-1 were examined using quantitative PCR. The invasiveness of oral cancer cells in the presence of conditioned media collected from ET-1 treated fibroblasts was determined using 2D Matrigel assays. KEY FINDINGS: Here we provide evidence that ET-1 increases the migration of oral fibroblasts and induces a more contractile phenotype which is not associated with changes in gene expression indicative of myofibroblast transdifferentiation. In addition we provide evidence that conditioned medium of ET-1-stimulated oral fibroblasts promotes invasion of OSCC cells in vitro. SIGNIFICANCE: In oral squamous cell carcinoma, a frequently fatal and increasingly common epithelial malignancy of the oral cavity, ET-1 is known to contribute to pro-migratory paracrine signalling between stromal fibroblasts and cancer cells. The ability of ET-1 to modulate the phenotype of human oral stromal fibroblasts, however, has not previously been reported. The findings presented here suggest that targeting the stromal endothelin system may be a viable and novel therapeutic strategy for invasive oral cancer.


Subject(s)
Carcinoma, Squamous Cell/pathology , Endothelin-1/metabolism , Fibroblasts/metabolism , Mouth Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Collagen/metabolism , Humans , Neoplasm Invasiveness , Paracrine Communication , Rats , Tail
11.
Biochim Biophys Acta ; 1751(1): 2-8, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16054014

ABSTRACT

In contrast to the relatively ubiquitous angiotensin-converting enzyme (ACE), expression of the mammalian ACE homologue, ACE2, was initially described in the heart, kidney and testis. ACE2 is a type I integral membrane protein with its active site domain exposed to the extracellular surface of endothelial cells and the renal tubular epithelium. Here ACE2 is poised to metabolise circulating peptides which may include angiotensin II, a potent vasoconstrictor and the product of angiotensin I cleavage by ACE. To this end, ACE2 may counterbalance the effects of ACE within the renin-angiotensin system (RAS). Indeed, ACE2 has been implicated in the regulation of heart and renal function where it is proposed to control the levels of angiotensin II relative to its hypotensive metabolite, angiotensin-(1-7). The recent solution of the structure of ACE2, and ACE, has provided new insight into the substrate and inhibitor profiles of these two key regulators of the RAS. As the complexity of this crucial pathway is unravelled, there is a growing interest in the therapeutic potential of agents that modulate the activity of ACE2.


Subject(s)
Carboxypeptidases/metabolism , Peptidyl-Dipeptidase A/metabolism , Amino Acid Sequence , Angiotensin-Converting Enzyme 2 , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Binding Sites , Drosophila Proteins/metabolism , Humans , Membrane Glycoproteins/metabolism , Metalloendopeptidases/metabolism , Receptors, Virus/metabolism , Renin-Angiotensin System/physiology , Severe acute respiratory syndrome-related coronavirus/metabolism , Substrate Specificity
12.
Br J Cancer ; 86(8): 1262-9, 2002 Apr 22.
Article in English | MEDLINE | ID: mdl-11953883

ABSTRACT

Healthy colonocytes derive 60-70% of their energy supply from short-chain fatty acids, particularly butyrate. Butyrate has profound effects on differentiation, proliferation and apoptosis of colonic epithelial cells by regulating expression of various genes associated with these processes. We have previously shown that butyrate is transported across the luminal membrane of the colonic epithelium via a monocarboxylate transporter, MCT1. In this paper, using immunohistochemistry and in situ hybridisation histochemistry, we have determined the profile of MCT1 protein and mRNA expression along the crypt to surface axis of healthy human colonic tissue. There is a gradient of MCT1 protein expression in the apical membrane of the cells along the crypt-surface axis rising to a peak in the surface epithelial cells. MCT1 mRNA is expressed along the crypt-surface axis and is most abundant in cells lining the crypt. Analysis of healthy colonic tissues and carcinomas using immunohistochemistry and Western blotting revealed a significant decline in the expression of MCT1 protein during transition from normality to malignancy. This was reflected in a corresponding reduction in MCT1 mRNA expression, as measured by Northern analysis. Carcinoma samples displaying reduced levels of MCT1 were found to express the high affinity glucose transporter, GLUT1, suggesting that there is a switch from butyrate to glucose as an energy source in colonic epithelia during transition to malignancy. The expression levels of MCT1 in association with GLUT1 could potentially be used as determinants of the malignant state of colonic tissue.


Subject(s)
Cell Transformation, Neoplastic , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Monocarboxylic Acid Transporters/metabolism , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , Symporters/metabolism , Aged , Aged, 80 and over , Blotting, Western , Colon/metabolism , Colon/pathology , Colonic Neoplasms/genetics , Female , Glucose Transporter Type 1 , Humans , Immunohistochemistry , In Situ Hybridization , Male , Middle Aged , Monocarboxylic Acid Transporters/genetics , Monosaccharide Transport Proteins/genetics , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Symporters/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...