Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Language
Publication year range
2.
Sci Rep ; 6: 34796, 2016 10 05.
Article in English | MEDLINE | ID: mdl-27703211

ABSTRACT

The risk of recurrence following radiation therapy remains high for a significant number of prostate cancer patients. The development of in vitro isogenic models of radioresistance through exposure to fractionated radiation is an increasingly used approach to investigate the mechanisms of radioresistance in cancer cells and help guide improvements in radiotherapy standards. We treated 22Rv1 prostate cancer cells with fractionated 2 Gy radiation to a cumulative total dose of 60 Gy. This process selected for 22Rv1-cells with increased clonogenic survival following subsequent radiation exposure but increased sensitivity to Docetaxel. This RR-22Rv1 cell line was enriched in S-phase cells, less susceptible to DNA damage, radiation-induced apoptosis and acquired enhanced migration potential, when compared to wild type and aged matched control 22Rv1 cells. The selection of radioresistant cancer cells during fractionated radiation therapy may have implications in the development and administration of future targeted therapy in conjunction with radiation therapy.


Subject(s)
Prostatic Neoplasms/genetics , Radiation Tolerance , S Phase , Cell Line, Tumor , Cell Survival/drug effects , Docetaxel , Dose Fractionation, Radiation , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/radiotherapy , Radiation Tolerance/drug effects , Reactive Oxygen Species , S Phase/drug effects , Taxoids/pharmacology
3.
BMC Cancer ; 16: 497, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27431913

ABSTRACT

BACKGROUND: Barrett's esophagus follows the classic step-wise progression of metaplasia-dysplasia-adenocarcinoma. While Barrett's esophagus is a leading known risk factor for esophageal adenocarcinoma, the pathogenesis of this disease sequence is poorly understood. Mitochondria are highly susceptible to mutations due to high levels of reactive oxygen species (ROS) coupled with low levels of DNA repair. The timing and levels of mitochondria instability and dysfunction across the Barrett's disease progression is under studied. METHODS: Using an in-vitro model representing the Barrett's esophagus disease sequence of normal squamous epithelium (HET1A), metaplasia (QH), dysplasia (Go), and esophageal adenocarcinoma (OE33), random mitochondrial mutations, deletions and surrogate markers of mitochondrial function were assessed. In-vivo and ex-vivo tissues were also assessed for instability profiles. RESULTS: Barrett's metaplastic cells demonstrated increased levels of ROS (p < 0.005) and increased levels of random mitochondrial mutations (p < 0.05) compared with all other stages of the Barrett's disease sequence in-vitro. Using patient in-vivo samples, Barrett's metaplasia tissue demonstrated significantly increased levels of random mitochondrial deletions (p = 0.043) compared with esophageal adenocarcinoma tissue, along with increased expression of cytoglobin (CYGB) (p < 0.05), a gene linked to oxidative stress, compared with all other points across the disease sequence. Using ex-vivo Barrett's metaplastic and matched normal patient tissue explants, higher levels of cytochrome c (p = 0.003), SMAC/Diablo (p = 0.008) and four inflammatory cytokines (all p values <0.05) were secreted from Barrett's metaplastic tissue compared with matched normal squamous epithelium. CONCLUSIONS: We have demonstrated that increased mitochondrial instability and markers of cellular and mitochondrial stress are early events in the Barrett's disease sequence.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/genetics , Gene Expression Regulation, Neoplastic , Metaplasia/genetics , Mitochondria/genetics , Mutation , Adenocarcinoma/metabolism , Barrett Esophagus/metabolism , Cell Line , Cell Line, Tumor , Cytochromes c/metabolism , Cytoglobin , Cytokines/metabolism , Disease Progression , Enzyme-Linked Immunosorbent Assay , Esophagus/metabolism , Esophagus/pathology , Globins/genetics , Globins/metabolism , Humans , Inflammation Mediators/metabolism , Metaplasia/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Risk Factors
4.
Clin. transl. oncol. (Print) ; 18(6): 632-640, jun. 2016. tab, ilus, graf
Article in English | IBECS | ID: ibc-152759

ABSTRACT

Purpose: Oesophageal adenocarcinoma is an exemplar model of obesity-associated cancer. Locally advanced disease is treated with neoadjuvant chemoradiotherapy, and survival rates are highest in patients demonstrating a pathological response following neoadjuvant therapy. Given that 55 % of oesophageal adenocarcinoma patients are obese, uncovering the effect of adipose tissue on radioresponse is clinically relevant. This study investigates if adipose tissue activates genomic instability events in radioresponsive (OE33P) and radioresistant (OE33R) oesophageal cancer cell lines and tumour samples. Methods: OE33R and OE33P were cultured with adiposeconditioned media derived from oesophageal adenocarcinoma patients (n = 10). Anaphase bridges, a marker of genomic instability, were enumerated in both cell lines following treatment with adipose media, and normalised to cell number. Genomic instability is regulated by the spindle assembly complex. Expression of two spindle assembly complex genes (MAD2L2, BUB1B) was assessed using qPCR, and validated in patient tumour specimens from viscerally obese (n = 46) and nonobese patients (n = 41). Results: Adipose-conditioned media increased anaphase bridging in OE33R (p < 0.0001), with a threefold increase in OE33R compared to OE33P (p < 0.01). Levels of anaphase bridges in OE33R cells correlated with visceral obesity status as measured by waist circumference (R = 0.709, p = 0.03) and visceral fat area (R = 0.794, p = 0.006). Adipose tissue altered expression of MAD2L2 in vitro. In vivo, MAD2L2 expression was higher in viscerally obese oesophageal adenocarcinoma patients compared with nonobese patients (p < 0.05). Conclusions: Anaphase bridge levels are influenced by obesity and radiosensitivity status in oesophageal adenocarcinoma. Furthermore, visceral adipose-conditioned media stimulates dysregulation of the spindle assembly complex in oesophageal adenocarcinoma patients


No disponible


Subject(s)
Humans , Male , Female , Esophageal Neoplasms/complications , Esophageal Neoplasms/radiotherapy , Adenocarcinoma/complications , Adenocarcinoma/diagnosis , Chemotherapy, Adjuvant/methods , Chemotherapy, Adjuvant , Genomic Instability , Genomic Instability/radiation effects , Obesity/complications , Obesity/pathology , Neoadjuvant Therapy , Anthropometry/methods , Adipose Tissue , Adipose Tissue/pathology
6.
Clin Transl Oncol ; 18(6): 632-40, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26474871

ABSTRACT

PURPOSE: Oesophageal adenocarcinoma is an exemplar model of obesity-associated cancer. Locally advanced disease is treated with neoadjuvant chemoradiotherapy, and survival rates are highest in patients demonstrating a pathological response following neoadjuvant therapy. Given that 55 % of oesophageal adenocarcinoma patients are obese, uncovering the effect of adipose tissue on radioresponse is clinically relevant. This study investigates if adipose tissue activates genomic instability events in radioresponsive (OE33P) and radioresistant (OE33R) oesophageal cancer cell lines and tumour samples. METHODS: OE33R and OE33P were cultured with adipose-conditioned media derived from oesophageal adenocarcinoma patients (n = 10). Anaphase bridges, a marker of genomic instability, were enumerated in both cell lines following treatment with adipose media, and normalised to cell number. Genomic instability is regulated by the spindle assembly complex. Expression of two spindle assembly complex genes (MAD2L2, BUB1B) was assessed using qPCR, and validated in patient tumour specimens from viscerally obese (n = 46) and nonobese patients (n = 41). RESULTS: Adipose-conditioned media increased anaphase bridging in OE33R (p < 0.0001), with a threefold increase in OE33R compared to OE33P (p < 0.01). Levels of anaphase bridges in OE33R cells correlated with visceral obesity status as measured by waist circumference (R = 0.709, p = 0.03) and visceral fat area (R = 0.794, p = 0.006). Adipose tissue altered expression of MAD2L2 in vitro. In vivo, MAD2L2 expression was higher in viscerally obese oesophageal adenocarcinoma patients compared with nonobese patients (p < 0.05). CONCLUSIONS: Anaphase bridge levels are influenced by obesity and radiosensitivity status in oesophageal adenocarcinoma. Furthermore, visceral adipose-conditioned media stimulates dysregulation of the spindle assembly complex in oesophageal adenocarcinoma patients.


Subject(s)
Adenocarcinoma/pathology , Cell Transformation, Neoplastic/genetics , Esophageal Neoplasms/pathology , Obesity, Abdominal/complications , Radiation Tolerance/genetics , Adenocarcinoma/genetics , Aged , Anaphase/genetics , Cell Cycle Proteins/analysis , Cell Cycle Proteins/biosynthesis , Cell Transformation, Neoplastic/pathology , Esophageal Neoplasms/genetics , Genomic Instability , Humans , M Phase Cell Cycle Checkpoints/genetics , Mad2 Proteins/analysis , Mad2 Proteins/biosynthesis , Male , Middle Aged , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/biosynthesis , Real-Time Polymerase Chain Reaction
7.
Cancer Lett ; 354(1): 122-31, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25107643

ABSTRACT

Contemporary clinical management of Barrett's oesophagus has highlighted the lack of accurate predictive markers of disease progression to oesophageal cancer. This study aims to examine alterations in mitochondrial energy metabolism profiles across the entire disease progression sequence in Barrett's oesophagus. An in-vitro model was used to screen 84 genes associated with mitochondrial energy metabolism. Three energy metabolism genes (ATP12A, COX4I2, COX8C) were significantly altered across the in-vitro Barrett's disease sequence. In-vivo validations across the Barrett's sequence demonstrated differential expression of these genes. Tissue microarrays demonstrated significant alterations in both epithelial and stromal oxidative phosphorylation (ATP5B and Hsp60) and glycolytic (PKM2 and GAPDH) protein markers across the in-vivo Barrett's sequence. Levels of ATP5B in sequential follow up surveillance biopsy material segregated Barrett's non progressors and progressors to HGD and cancer. Utilising the Seahorse XF24 flux analyser, in-vitro Barrett's and adenocarcinoma cells exhibited altered levels of various oxidative parameters. We show for the first time that mitochondrial energy metabolism is differentially altered across the metaplasia-dysplasia-adenocarcinoma sequence and that oxidative phosphorylation profiles have predictive value in segregating Barrett's non progressors and progressors to adenocarcinoma.


Subject(s)
Adenocarcinoma/metabolism , Barrett Esophagus/metabolism , Gene Expression Regulation, Neoplastic , Mitochondria/metabolism , Barrett Esophagus/pathology , Biopsy , Cell Line, Tumor , Disease Progression , Electron Transport Complex IV/metabolism , Energy Metabolism , Gene Expression Profiling , Glycolysis , H(+)-K(+)-Exchanging ATPase/metabolism , Humans , Metaplasia/metabolism , Oligonucleotide Array Sequence Analysis , Oxygen/chemistry , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL
...