Carol Beesley
University of Liverpool
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Carol Beesley.
Oncogene | 2003
Janet Adamson; Elwin A. Morgan; Carol Beesley; Yongqiang Mei; Christopher S. Foster; Hiroshi Fujii; Philip S. Rudland; Paul H. Smith; Youqiang Ke
The expression of cutaneous fatty acid-binding protein (C-FABP) in prostate tissues was examined by immunohistochemistry. Among the 76 cases, all seven (100%) normal tissues were unstained. Of the 35 benign prostatic hyperplasia (BPH), 25 (71.4%) specimens were unstained and 10 (28.6%) were stained positively. For the 34 prostatic carcinomas, the C-FABP expression was remarkably increased: 25 (73.5%) samples stained positively, and only nine (26.5%) were unstained. Transfection of a vector expressing an antisense C-FABP transcript into the PC-3M prostatic cancer cells yielded two transfectant lines: PC-3M-CFABP-1 and PC-3M-CFABP-3, producing, respectively, a 3.8- and a 6.9-fold reduction in C-FABP levels. Comparing with the control transfectants, the in vitro invasiveness of both PC-3M-CFABP-1 and PC-3M-CFABP-3 was significantly reduced. When tested in nude mouse, the average size of tumours produced by PC-3M-CFABP-1 and by PC-3M-CFABP-3 was reduced by 2.9- and 4.2-fold respectively, in comparison with that of tumours produced by the control transfectants. Analysis showed that the decreased vascular endothelial growth factor (VEGF) and microvessel densities in the tumours were associated with the reduced C-FABP. These data show that C-FABP is increased in prostatic carcinoma cells and suppression of its expression can significantly inhibit the tumorigenicity, probably by reducing the expression of VEGF.
Clinical Cancer Research | 2006
Alix Bee; Youqianq Ke; Shiva S. Forootan; Ke Lin; Carol Beesley; Sharon E. Forrest; Christopher S. Foster
Microquantity differential display analysis of gene expression profiles between benign (PNT2) and malignant (PC3M) human prostate cell lines identified the gene encoding ribosomal protein L19 (RPL19) to be overexpressed in the malignant cells. Northern blot hybridization analysis done on a wide range of human cell lines and tissues confirmed the level of RPL19 mRNA to be 5-fold higher in malignant cell lines and 8-fold higher in malignant tissues, when compared with their benign counterparts. Analysis of RPL19 mRNA expression by in situ hybridization revealed a significant increase of RPL19 expression in a substantial number of prostate cancers. All of the eight normal prostatic tissues were unstained (100%). Of 32 benign prostatic hyperplasia (BPH) tissues, 15 (46.9%) were unstained, 9 (28.1%) stained weakly, and 8 (25%) stained moderately. Among 87 carcinomas, only 7 (8.1%) were unstained, whereas 22 (25.2%) stained weakly, 21 (24.1%) stained moderately, and 37 (42.61%) stained strongly. The intensity of staining of the malignant specimens was significantly higher than that of normal and BPH specimens (χ2: n = 127, P < 0.001). Gleason scores of the carcinomas correlated with RPL19 expression (χ2: n = 87, P < 0.001). Kaplan-Meier survival analysis confirmed increased RPL19 expression to be highly predictive of shorter patient survival (P < 0.05), revealing RPL19 to be a sensitive predictor of prostate cancer progression. Expression of this protein could be a valuable marker in prostate cancer diagnosis and patient management.
Clinical Cancer Research | 2008
Laleh Kamalian; John R. Gosney; Shiva S. Forootan; Christopher S. Foster; Zheng Z. Bao; Carol Beesley; Youqiang Ke
Purpose: To study the molecular pathology of human small cell lung cancer (SCLC), molecular biology approaches were used to identify genes involved in malignant progression of the cancer cells. Experimental Design: Microquantity differential display was used initially to identify genes expressed differentially between normal and malignant cell lines. The differences were verified by Western blot. Immunohistochemical analysis was done on paired normal and malignant lung tissues and on tissues taken by biopsy to assess the expression status of candidate genes and their prognostic significance. Results:Inhibitor of DNA/differentiation (Id)1 gene was up-regulated in SCLC cells. Levels of Id1 in 8 of 10 cell lines were increased by 1.7- to 21.4-fold when compared with the benign cells. A similar increase was also found in levels of Id2 and Id3. On 26 pairs of lung tissues, all four Id proteins were significantly (Wilcoxon Signed Rank Test, P < 0.001-0.005) overexpressed in cytoplasm of the malignant cells. In nuclei of SCLC cells, Id1 expression was significantly reduced, whereas the levels of Id2, Id3, and Id4 were significantly (Wilcoxon Signed Rank Test, P < 0.001) increased. Immunohistochemical staining on biopsy specimens showed that the increased expression of Id2 in cytoplasm of cancer cells, not the other three proteins, was significantly associated with the increased survival of SCLC patients. Conclusion: Changed expression profiles of Id proteins may play important roles in malignant progression of SCLC, and the increased Id2 in cytoplasm is a novel prognostic factor to predict the patient outcomes.
Genes & Cancer | 2010
Sheng Yao; Alix Bee; Daniel Brewer; Andrew Dodson; Carol Beesley; Youqiang Ke; Laurence Ambroisine; Gabrielle Fisher; Heinrich Møller; Tim Dickinson; Patricia Gerard; Lu-Yu Lian; Janet M. Risk; Brian Lane; Paul Smith; Victor E. Reuter; Daniel M. Berney; Christine Gosden; Peter T. Scardino; Jack Cuzick; M.B.A. Djamgoz; Colin S. Cooper; Christopher S. Foster
We show protein kinase C-zeta (PKC-ζ) to be a novel predictive biomarker for survival from prostate cancer (P < 0.001). We also confirm that transcription of the PRKC-ζ gene is crucial to the malignant phenotype of human prostate cancer. Following siRNA silencing of PRKC-ζ in PC3-M prostate cancer cells, stable transfectant cell line si-PRKC-ζ-PC3-M(T1-6) is phenotypically nonmalignant in vitro and in vivo. Genome-wide expression analysis identified 373 genes to be differentially expressed in the knockdown cells and 4 key gene networks to be significantly perturbed during phenotype modulation. Functional interconnection between some of the modulated genes is revealed, although these may be within different regulatory pathways, emphasizing the complexity of their mutual interdependence. Genes with altered expression following PRKC-ζ knockdown include HSPB1, RAD51, and ID1 that we have previously described to be critical in prostatic malignancy. Because expression of PRKC-ζ is functionally involved in promoting the malignant phenotype, we propose PKC-ζ as a novel and biologically relevant target for therapeutic intervention in prostate cancer.
PLOS ONE | 2011
Alix Bee; Daniel Brewer; Carol Beesley; Andrew Dodson; Shiva S. Forootan; Timothy Dickinson; Patricia Gerard; Brian Lane; Sheng Yao; Colin S. Cooper; M.B.A. Djamgoz; Christine Gosden; Youqiang Ke; Christopher S. Foster
We provide novel functional data that posttranscriptional silencing of gene RPL19 using RNAi not only abrogates the malignant phenotype of PC-3M prostate cancer cells but is selective with respect to transcription and translation of other genes. Reducing RPL19 transcription modulates a subset of genes, evidenced by gene expression array analysis and Western blotting, but does not compromise cell proliferation or apoptosis in-vitro. However, growth of xenografted tumors containing the knocked-down RPL19 in-vivo is significantly reduced. Analysis of the modulated genes reveals induction of the non-malignant phenotype principally to involve perturbation of networks of transcription factors and cellular adhesion genes. The data provide evidence that extra-ribosomal regulatory functions of RPL19, beyond protein synthesis, are critical regulators of cellular phenotype. Targeting key members of affected networks identified by gene expression analysis raises the possibility of therapeutically stabilizing a benign phenotype generated by modulating the expression of an individual gene and thereafter constraining a malignant phenotype while leaving non-malignant tissues unaffected.
Prostate Cancer and Prostatic Diseases | 2013
Nataša Vasiljević; Amar Ahmad; Carol Beesley; Mangesh A. Thorat; Gabrielle Fisher; D. Berney; Henrik Møller; Yongwei Yu; Y-J Lu; Jack Cuzick; Christopher S. Foster; Attila T. Lorincz
Background:Heat shock protein 27 (Hsp-27) encoded by gene HSPB1 is a critical regulator of the behavioral phenotype of human prostate cancer (PCa) cells, enhanced expression being associated with highly aggressive disease and poor clinical outcome. In contrast, the protein is not expressed in PCas of low malignant potential. To gain insight into the mechanism regulating its expression, we tested the hypothesis that differential methylation of CpG islands within HSPB1 controls transcription and subsequent translation of the gene.Methods:We studied prostate epithelial cell lines and tissue biopsies, including 59 BPH and 415 PCas, of which 367 were a cohort of men with up to 20 years of follow-up. Methylation across the gene (DNA methylation (DNAme)) was assayed by pyrosequencing. Hsp-27 expression was assessed by western blot and immunohistochemistry.Results:In cancer tissues, methylation increased in a 3′ direction (P<0.0001) whereas in benign hyperplasia methylation was constantly below 5%, a cutoff giving a specificity of 100% and sensitivity of 50%. Although methylation of the promoter region was significantly discriminating between benign and malignant prostatic epithelia, it compared poorly with methylation of the first intron. The prognostic value of HSPB1 DNAme was confirmed by both univariate (hazard ratio 1.77 per 50% increment, P=0.02) and multivariate models. Interaction between HSPB1 methylation and Gleason score revealed high DNAme to be a reliable prognostic marker of poor outcome in men with low Gleason score (P=0.014).Conclusions:Our data indicate CpG methylation of the first HSPB1 intron to be an important biomarker that identifies aggressive PCas otherwise regarded as low risk by current clinical criteria but that, biologically, require immediate active management.
Urology | 1998
Paul Smith; Nick Rhodes; Carol Beesley; Youqiang Ke; Christopher S. Foster
OBJECTIVES To evaluate the response of prostatic stromal cells in vitro to the action of the agonist norepinephrine and to examine the role of cell density in this response. METHODS Stromal cells isolated from transurethral chippings of patients with benign prostatic hyperplasia (BPH) were seeded onto tissue culture dishes either at high density (9 x 10(3) cells/cm2) or at low density (1.5 x 10(3) cells/cm2). Norepinephrine was added at concentrations in the range of 2.5 to 50 microM. Cells were harvested at the moment of confluence, labeled with monoclonal antibodies to four cytoskeletal proteins, and analyzed by flow cytometry. RESULTS Sparsely plated stromal cells showed a consistent biphasic response in which a small fall in immunofluorescence occurred in the range of 5 to 15 microM norepinephrine but was thereafter followed by a progressive rise in fluorescence to 50 microM, indicating increased expression of smooth-muscle-associated cytoskeletal proteins. The shape of flow-cytometric frequency-distribution histograms for smooth-muscle myosin, desmin, and talin suggested that all mesenchymal cells in the stromal cultures were similarly modulated by norepinephrine. However, the effect on smooth-muscle actin was different in that a subpopulation of hyperreactive cells was identified. Densely plated stromal cells did not show a similar biphasic response to norepinephrine but instead demonstrated an overall downward trend, indicating a progressively diminished expression of these cytoskeletal proteins. CONCLUSIONS Norepinephrine stimulation directly modulates BPH-derived prostatic stromal cells toward a differentiated smooth-muscle phenotype as evidenced by increased expression of cytoskeletal proteins. The effect of norepinephrine on cultures is cell-density-dependent, suggesting that intercellular communication is an important factor in coordinating the differentiation responses. This study has revealed a specific interaction between physical and humoral stimuli, which influences in part the phenotype of prostatic stromal cells. Such interaction is likely to determine the development of clinical BPH, and also the response of any individuals following therapeutic intervention using selective alpha-adrenergic blockade.
British Journal of Cancer | 2012
Sheng Yao; Ireland Sj; Alix Bee; Carol Beesley; Shiva S. Forootan; Andrew Dodson; Dickinson T; Patricia Gerard; Lian Ly; Janet M. Risk; Paul Smith; Mohammed I. Malki; Youqiang Ke; Colin S. Cooper; Christine Gosden; Christopher S. Foster
Background:Previously, using gene-knockdown techniques together with genome expression array analysis, we showed the gene protein Kinase C (PKC)-zeta (PRKCZ) to mediate the malignant phenotype of human prostate cancer. However, according to NCBI, the gene has undergone several major iterations. Therefore, to understand the relationship between its structure and biological activities, we have analysed its expressed sequence in prostate cancer cell lines and tissues.Methods:Transcriptome-walking and targeted PCR were used to sequence the mRNA transcribed from PRKCZ. Hydropathy analysis was employed to analyse the hypothetical protein sequence subsequently translated and to identify an appropriate epitope to generate a specific monoclonal antibody.Results:A novel sequence was identified within the 3′-terminal domain of human PRKCZ that, in prostate cancer cell lines and tissues, is expressed during transcription and thereafter translated into protein (designated PKC-ζ-PrC) independent of conventional PKC-ζ-a. The monoclonal antibody detected expression of this 96 kD protein only within malignant prostatic epithelium.Interpretation:Transcription and translation of this gene sequence, including previous intronic sequences, generates a novel specific biomarker of human prostate cancer. The presence of catalytic domains characteristic of classic PKC-β and atypical PKC-ι within PKC-ζ-PrC provides a potential mechanism for this PRKCZ variant to modulate the malignant prostatic phenotype out-with normal cell-regulatory control.
European Respiratory Journal | 1997
Gosney; J Peers; Carol Beesley; E Gradwell
It has been suggested by some studies of human and animal lungs that the products of pulmonary endocrine cells, particularly gastrin-releasing peptide, might play a role in fibrogenesis, but more recent detailed studies of fibrotic human lungs have failed to confirm this. We have made a detailed quantitative examination of a series of fibrotic human lungs to see if we could determine whether there was any relationship between endocrine cells and fibrosis. Using immunocytochemistry, we investigated the morphology, content, distribution and number of pulmonary endocrine cells in 15 pairs of fibrotic lungs from coal miners, and compared their features with those of equivalent cells in age-matched controls. Proliferation of endocrine cells was seen in the lungs of just two miners, in which it was focal and associated with acute bronchitis and bronchopneumonia. There was no difference between the miners and controls in the appearance (mostly solitary cells), content (predominantly gastrin-releasing peptide and calcitonin), distribution (mainly in small bronchi and bronchioles), or number (4.5 vs 4.1 cells per 10,000 epithelial cells, respectively) of endocrine cells. It seems unlikely that the substances secreted by these cells play any role in stimulating fibrosis in human lungs, but rather that they have a function in the inflammatory response to pulmonary injury.
Journal of the National Cancer Institute | 2002
Chun Jing; Manal Abd El-Ghany; Carol Beesley; Christopher S. Foster; Philip S. Rudland; Paul Smith; Youqiang Ke