Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Asia Pac J Clin Oncol ; 17(1): 131-138, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32885561

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is rising in incidence in young adults, and this observation is currently unexplained. We investigated whether having a personal history of type 2 diabetes mellitus (T2D) was a potential risk factor for young-onset colorectal cancer (YOCRC). METHODS: The South Australian Young Onset (SAYO) CRC study is a series of young adults with CRC below age 55. Ninety unrelated YOCRC cases were recruited to the study. Personal history and detailed family history of T2D were obtained at face-to-face interview and confirmed from medical records. Whole exome sequencing was conducted on germline DNA from each CRC case. Controls for personal history studies of T2D were 240 patients with proven clear colonoscopies and no known CRC predispositions. RESULTS: The median age of YOCRC cases was 44 years (18-54) and of controls was 45 years (18-54), and 53% of both cases and controls were females (P = 0.99). Left-sided (distal) CRC was seen in 67/89 (75%) of cases. A personal history of T2D was confirmed in 17/90 (19%) YOCRC patients compared with controls (12/240, 5%; P < 0.001; odds ratio = 4.4; 95% confidence interval, 2.0-9.7). YOCRC patients frequently reported at least one first-degree relative with T2D (32/85, 38%). Ten of 87 (12%) of YOCRC cases had CRC-related pathogenic germline variants, however, no pathogenic variants in familial diabetes-associated genes were seen. CONCLUSIONS: Though the mechanism remains unclear, our observations suggest that there is enrichment for personal history of T2D in YOCRC patients. IMPACT: A diagnosis of T2D could therefore potentially identify a subset of young adults at increased risk for CRC and in whom early screening might be appropriate.


Subject(s)
Colorectal Neoplasms/etiology , Diabetes Mellitus, Type 2/complications , Adolescent , Adult , Age of Onset , Australia , Colorectal Neoplasms/pathology , Female , Genotype , Humans , Male , Middle Aged , Risk Factors , Young Adult
2.
Dig Dis Sci ; 64(1): 76-83, 2019 01.
Article in English | MEDLINE | ID: mdl-30382540

ABSTRACT

BACKGROUND: Wnt-ß-catenin signaling is essential for homeostasis of intestinal stem cells in mice and is thought to promote intestinal crypt fission. AIMS: The aim of this study was to investigate Wnt-ß-catenin signaling in intestinal crypts of human infants. METHODS: Duodenal biopsies from nine infants (mean, range 0.9 years, 0.3-2 years) and 11 adults (mean, range 43 years, 34-71 years) were collected endoscopically. Active ß-catenin signaling was assessed by cytoplasmic and nuclear ß-catenin, nuclear c-Myc, and cytoplasmic Axin-2 expression in the base of crypts. Tissues were stained by an immunoperoxidase staining technique and quantified as pixel energy using cumulative signal analysis. Data were expressed as mean ± SD and significance assessed by Student's t test. RESULTS: Crypt fission was significantly higher in infants compared to adults (16 ± 8.6% versus 0.7 ± 0.6%, respectively, p < 0.0001). Expression of cytoplasmic and nuclear ß-catenin was 1.8-fold (p < 0.0001) and 2.9-fold (p < 0.0001) higher in infants, respectively, while cytoplasmic Axin-2 was 3.1-fold (p < 0.0001) increased in infants. c-Myc expression was not significantly different between infants and adults. Expression was absent in Paneth cells but present in the transit amplifying zone of crypts. Crypt base columnar cells, which were intercalated between Paneth cells, expressed c-Myc. CONCLUSIONS: Wnt-ß-catenin signaling was active in crypt base columnar cells (i.e., intestinal stem cells) in human infants. This signaling could promote crypt fission during infancy. Wnt-ß-catenin signaling likely acts in concert with other pathways to promote postnatal growth.


Subject(s)
Duodenum/chemistry , Intestinal Mucosa/chemistry , Wnt Signaling Pathway , beta Catenin/analysis , Adult , Age Factors , Aged , Axin Protein/analysis , Duodenum/growth & development , Female , Humans , Infant , Intestinal Mucosa/growth & development , Male , Middle Aged , Paneth Cells/chemistry , Proto-Oncogene Proteins c-myc/analysis , Stem Cells/chemistry
3.
Oncotarget ; 9(27): 19100-19114, 2018 Apr 10.
Article in English | MEDLINE | ID: mdl-29721186

ABSTRACT

Fibroblasts express androgen receptor (AR) in the normal prostate and during prostate cancer development. We have reported that loss of AR expression in prostate cancer-associated fibroblasts is a poor prognostic indicator. Here we report outcomes of direct and indirect co-cultures of immortalised AR-positive (PShTert-AR) or AR-negative (PShTert) myofibroblasts with prostate cancer cells. In the initial co-cultures the AR-negative PC3 cell line was used so AR expression and signalling were restricted to the myofibroblasts. In both direct and indirect co-culture with PShTert-AR myofibroblasts, paracrine signalling to the PC3 cells slowed proliferation and induced apoptosis. In contrast, PC3 cells proliferated with PShTert myofibroblasts irrespective of the co-culture method. In direct co-culture PC3 cells induced apoptosis in and destroyed PShTerts by direct signalling. Similar results were seen in direct co-cultures with AR-negative DU145 and AR-positive LNCaP and C4-2B prostate cancer cell lines. The AR ligand 5α-dihydrotestosterone (DHT) inhibited the proliferation of the PShTert-AR myofibroblasts, thereby reducing the extent of their inhibitory effect on cancer cell growth. These results suggest loss of stromal AR would favour prostate cancer cell growth in vivo, providing an explanation for the clinical observation that reduced stromal AR is associated with a poorer outcome.

4.
Dig Dis Sci ; 62(12): 3402-3414, 2017 12.
Article in English | MEDLINE | ID: mdl-29052817

ABSTRACT

BACKGROUND: We showed previously that nuclear localization of the androgen receptor (AR) and expression of the androgen-responsive gene FK506-binding protein 5 (FKBP5) in esophageal adenocarcinoma (EAC) tissues were associated with decreased patient survival, suggesting a role for androgens in this cancer. AIM: To investigate the effect of the AR ligand 5α-dihydrotestosterone (DHT) on AR-expressing EAC cell lines in vitro. METHODS AND RESULTS: In tissue resection specimens from EAC patients, FKBP5 expression was positively associated with proliferation as measured by Ki-67 expression. We stably transduced AR into three AR-negative EAC cell lines, OE33, JH-EsoAd1, and OE19, to investigate androgen signaling in vitro. In the AR-expressing cell lines, 10 nM DHT, the concentration typically used to study AR signaling, induced changes in the expression of androgen-responsive genes and inhibited proliferation by inducing cell cycle arrest and senescence. At lower DHT concentrations near the half maximal inhibitory concentration (IC50), the AR-expressing cell lines proliferated and there were changes in the expression of androgen-responsive genes. In direct co-culture with cancer-associated fibroblast-like PShTert myofibroblasts, 10 nM DHT induced changes in the expression of androgen-responsive genes but did not inhibit proliferation. CONCLUSIONS: This is the first study to show that EAC cell lines respond to androgen in vitro. Proliferation together with the expression of androgen-responsive genes was dependent on the concentration of DHT, or the presence of a permissive microenvironment, consistent with observations in the tissues. These findings are consistent with a role for androgen signaling in EAC.


Subject(s)
Adenocarcinoma/metabolism , Androgens/metabolism , Esophageal Neoplasms/metabolism , Receptors, Androgen/metabolism , Tacrolimus Binding Proteins/metabolism , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cellular Senescence , Coculture Techniques , Dihydrotestosterone , Fibroblasts/physiology , Gene Expression Regulation , Humans
5.
Sci Rep ; 7(1): 3368, 2017 06 13.
Article in English | MEDLINE | ID: mdl-28611465

ABSTRACT

Oesophageal adenocarcinoma (OAC) is increasing in incidence and has a poor prognosis. Tumour derived fibroblasts (TDFs) differ functionally from normal fibroblasts (NDFs), and play a pivotal role in cancer. Many of the differences persist through subculture. We measured the DNA methylation profiles of 10 TDFs from OAC with 12 NDF from normal oesophageal mucosa using Infinium HumanMethylation450 Beadchips and found they differed in multidimensional scaling analysis. We identified 4,856 differentially methylated CpGs (DMCs, adjusted p < 0.01 and absolute difference in average ß-value > 0.15), of which 3,243 (66.8%) were hypomethylated in TDFs compared to NDFs. Hypermethylated DMCs were enriched at transcription start sites (TSSs) and in CpG islands, and depleted in transcriptional enhancers. Gene ontology analysis of genes with DMCs at TSSs revealed an enrichment of genes involved in development, morphogenesis, migration, adhesion, regulation of processes and response to stimuli. Alpha-smooth muscle actin (α-SMA) is a marker of activated fibroblasts and a poor prognostic indicator in OAC. Hypomethylated DMCs were observed at the TSS of transcript variant 2 of α-SMA, which correlated with an increase in α-SMA protein expression. These data suggest that DNA methylation may contribute to the maintenance of the TDF phenotype.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , DNA Methylation , Esophageal Neoplasms/genetics , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Genome, Human , Adenocarcinoma/pathology , Aged , Aged, 80 and over , Cells, Cultured , CpG Islands , Esophageal Neoplasms/pathology , Female , Fibroblasts/pathology , Humans , Male , Middle Aged , Transcription Initiation Site
6.
Anticancer Drugs ; 27(9): 848-53, 2016 10.
Article in English | MEDLINE | ID: mdl-27400374

ABSTRACT

This study examined the in-vivo effect of the NSAID celecoxib on DNA methylation in the promoter region of the tumor-suppressor genes cadherin 13, tissue factor pathway inhibitor 12, and follistatin-like protein 1, and on apoptosis, in esophageal squamous cell carcinoma (ESCC). Forty-five patients who underwent an esophagectomy for ESCC were allocated to either a treatment group (n=22) or a control group (n=23). Patients in the treatment group were administered 800 mg/day of celecoxib for 14 days before surgery. Patients in the control group did not take any type of NSAID. Biopsies of the tumor were collected before surgery and tissue from the resection specimens after surgery. Methylation-specific PCR was used to measure DNA methylation and apoptosis was measured by flow cytometry. There was no difference in the proportion of patients with methylation for each of the genes between the patient groups before treatment. In those patients with pretreatment methylation, there was a significant reduction in the proportion with methylation and a significant increase in the corresponding messenger RNA expression after treatment with celecoxib. In those tissues in which there was a reduction in methylation following celecoxib treatment, there was a significant increase in the percentage of apoptotic cells, but not in the tissues with no change in methylation. In ESCC, in-vivo treatment with celecoxib is associated with a reduction in DNA methylation and increase in messenger RNA expression of tumor-suppressor genes, and increases in apoptosis.


Subject(s)
Cadherins/genetics , Carcinoma, Squamous Cell/drug therapy , Celecoxib/administration & dosage , DNA Methylation/drug effects , Esophageal Neoplasms/drug therapy , Follistatin-Related Proteins/genetics , Glycoproteins/genetics , Adult , Aged , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/surgery , Cyclooxygenase 2 Inhibitors/administration & dosage , Esophageal Neoplasms/genetics , Esophageal Neoplasms/surgery , Female , Gene Expression/drug effects , Genes, Tumor Suppressor/drug effects , Humans , Male , Middle Aged , Promoter Regions, Genetic/drug effects
7.
Eur J Pharmacol ; 774: 144-9, 2016 Mar 05.
Article in English | MEDLINE | ID: mdl-26852958

ABSTRACT

Mammalian tachykinins are a family of neuropeptides which are potent modulators of smooth muscle function with a significant contractile effect on human smooth muscle preparations. Tachykinins act via three distinct G protein-coupled neurokinin (NK) receptors, NK1, NK2 and NK3, coded by the genes TACR1, TACR2 and TACR3 respectively. The purpose of this paper was to measure the mRNA and protein expression of these receptors and their isoforms in the clasp and sling fibers of the human lower esophageal sphincter complex and circular muscle from the adjacent distal esophagus and proximal stomach. We found differences in expression between the different receptors within these muscle types, but the rank order of the receptor expression did not differ between the different muscle types. The rank order of the mRNA expression was TACR2 (α isoform)>TACR2 (ß isoform)>TACR1 (short isoform)>TACR1 (long isoform)>TACR3. The rank order of the protein expression was NK2>NK1>NK3. This is the first report of the measurement of the transcript and protein expression of the tachykinin receptors and their isoforms in the muscles of the human lower esophageal sphincter complex. The results provide evidence that the tachykinin receptors could contribute to the regulation of the human lower esophageal sphincter, particularly the TACR2 α isoform which encodes the functional isoform of the tachykinin NK2 receptor was the most highly expressed of the tachykinin receptors in the muscles associated with the lower esophageal sphincter.


Subject(s)
Esophageal Sphincter, Lower/metabolism , Gene Expression Regulation , Receptors, Tachykinin/genetics , Receptors, Tachykinin/metabolism , Aged , Female , Humans , Male , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tachykinins/metabolism
8.
Dig Dis Sci ; 61(2): 433-43, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26467701

ABSTRACT

BACKGROUND: Esophageal adenocarcinoma is a male-dominant disease, but the role of androgens is unclear. AIMS: To examine the expression and clinical correlates of the androgen receptor (AR) and the androgen-responsive gene FK506-binding protein 5 (FKBP5) in esophageal adenocarcinoma. METHODS: Expression of AR and FKBP5 was determined by immunohistochemistry. The effect of the AR ligand 5α-dihydrotestosterone (DHT) on the expression of a panel of androgen-responsive genes was measured in AR-positive and AR-negative esophageal adenocarcinoma cell lines. Correlations in expression between androgen-responsive genes were analyzed in an independent cohort of esophageal adenocarcinoma tissues. RESULTS: There was AR staining in 75 of 77 cases (97 %), and FKBP5 staining in 49 (64 %), all of which had nuclear AR. Nuclear AR with FKBP5 expression was associated with decreased median survival (451 vs. 2800 days) and was an independent prognostic indicator (HR 2.894, 95 % CI 1.396­6.002, p = 0.0043) in multivariable Cox proportional hazards models. DHT induced a significant increase in expression of the androgen-responsive genes FKBP5, HMOX1, FBXO32, VEGFA, WNT5A, and KLK3 only in AR-positive cells in vitro. Significant correlations in expression were observed between these androgen-responsive genes in an independent cohort of esophageal adenocarcinoma tissues. CONCLUSION: Nuclear AR and expression of FKBP5 is associated with decreased survival in esophageal adenocarcinoma.


Subject(s)
Adenocarcinoma/metabolism , Esophageal Neoplasms/metabolism , Genetic Predisposition to Disease , Receptors, Androgen/metabolism , Tacrolimus Binding Proteins/metabolism , Adenocarcinoma/genetics , Cell Line, Tumor , Esophageal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , Prognosis , Receptors, Androgen/genetics , Tacrolimus Binding Proteins/genetics
10.
Oncotarget ; 6(18): 16135-50, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-25965833

ABSTRACT

Androgen receptor (AR) signaling in stromal cells is important in prostate cancer, yet the mechanisms underpinning stromal AR contribution to disease development and progression remain unclear. Using patient-matched benign and malignant prostate samples, we show a significant association between low AR levels in cancer associated stroma and increased prostate cancer-related death at one, three and five years post-diganosis, and in tissue recombination models with primary prostate cancer cells that low stromal AR decreases castration-induced apoptosis. AR-regulation was found to be different in primary human fibroblasts isolated from adjacent to cancerous and non-cancerous prostate epithelia, and to represent altered activation of myofibroblast pathways involved in cell cycle, adhesion, migration, and the extracellular matrix (ECM). Without AR signaling, the fibroblast-derived ECM loses the capacity to promote attachment of both myofibroblasts and cancer cells, is less able to prevent cell-matrix disruption, and is less likely to impede cancer cell invasion. AR signaling in prostate cancer stroma appears therefore to alter patient outcome by maintaining an ECM microenvironment inhibitory to cancer cell invasion. This paper provides comprehensive insight into AR signaling in the non-epithelial prostate microenvironment, and a resource from which the prognostic and therapeutic implications of stromal AR levels can be further explored.


Subject(s)
Myofibroblasts/pathology , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Stromal Cells/pathology , Tumor Microenvironment , Aged , Aged, 80 and over , Androgens/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Case-Control Studies , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , Follow-Up Studies , Humans , Immunoenzyme Techniques , Male , Middle Aged , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Neoplasm Grading , Neoplasm Invasiveness , Orchiectomy , Prognosis , Prostatic Hyperplasia/drug therapy , Prostatic Hyperplasia/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Androgen/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Stromal Cells/drug effects , Stromal Cells/metabolism , Tissue Array Analysis , Tumor Cells, Cultured
11.
World J Gastroenterol ; 20(21): 6554-9, 2014 Jun 07.
Article in English | MEDLINE | ID: mdl-24914377

ABSTRACT

AIM: To compare the binding of cholecystokinin (CCK)-8 to CCK receptors in sling and clasp fibers of the human lower esophageal sphincter. METHODS: Esophageal sling and clasp fibers were isolated from eight esophagectomy specimens, resected for squamous cell carcinoma in the upper two thirds of the esophagus, which had been maintained in oxygenated Kreb's solution. Western blot was used to measure CCK-A and CCK-B receptor subtypes in the two muscles. A radioligand binding assay was used to determine the binding parameters of (3)H-CCK-8S to the CCK receptor subtypes. The specificity of binding was determined by the addition of proglumide, which blocks the binding of CCK to both receptor subtypes. RESULTS: There was no significant difference between the sling and clasp fibers of the human lower esophageal sphincter in the amount of CCK-A [integrated optical density (IOD) value: 22.65 ± 0.642 vs 22.328 ± 1.042, P = 0.806] or CCK-B receptor protein (IOD value: 13.20 ± 0.423 vs 12.45 ± 0.294, P = 0.224) as measured by Western blot. The maximum binding of radio-labeled CCK-8S was higher in the sling fibers than in the clasp fibers (595.75 ± 3.231 cpm vs 500.000 ± 10.087 cpm, P < 0.001) and dissociation constant was lower (K(d): 1.437 ± 0.024 nmol/L vs 1.671 ± 0.024 nmol/L, P < 0.001). The IC50 of the receptor specific antagonists were lower for the CCK-A receptors than for the CCK-B (P < 0.01). CONCLUSION: CCK binding modulates the contractile function of the lower esophageal sphincter through differential binding to the CCK-A receptor on the sling and clasp fibers.


Subject(s)
Esophageal Sphincter, Lower/pathology , Esophagogastric Junction/pathology , Gene Expression Regulation , Receptor, Cholecystokinin A/metabolism , Receptor, Cholecystokinin B/metabolism , Sincalide/metabolism , Aged , Blotting, Western , Esophagectomy , Esophagogastric Junction/metabolism , Esophagus/pathology , Female , Humans , Inhibitory Concentration 50 , Isotonic Solutions , Male , Middle Aged , Proglumide/chemistry
12.
PLoS One ; 8(9): e75517, 2013.
Article in English | MEDLINE | ID: mdl-24086551

ABSTRACT

The miR-200b~200a~429 gene cluster is a key regulator of EMT and cancer metastasis, however the transcription-based mechanisms controlling its expression during this process are not well understood. We have analyzed the miR-200b~200a~429 locus for epigenetic modifications in breast epithelial and mesenchymal cell lines using chromatin immunoprecipitation assays and DNA methylation analysis. We discovered a novel enhancer located approximately 5.1kb upstream of the miR-200b~200a~429 transcriptional start site. This region was associated with the active enhancer chromatin signature comprising H3K4me1, H3K27ac, RNA polymerase II and CpG dinucleotide hypomethylation. Luciferase reporter assays revealed the upstream enhancer stimulated the transcription of the miR-200b~200a~429 minimal promoter region approximately 27-fold in breast epithelial cells. Furthermore, we found that a region of the enhancer was transcribed, producing a short, GC-rich, mainly nuclear, non-polyadenylated RNA transcript designated miR-200b eRNA. Over-expression of miR-200b eRNA had little effect on miR-200b~200a~429 promoter activity and its production did not correlate with miR-200b~200a~429 gene expression. While additional investigations of miR-200b eRNA function will be necessary, it is possible that miR-200b eRNA may be involved in the regulation of miR-200b~200a~429 gene expression and silencing. Taken together, these findings reveal the presence of a novel enhancer, which contributes to miR-200b~200a~429 transcriptional regulation in epithelial cells.


Subject(s)
Breast Neoplasms/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Cell Line, Tumor , Chromatin/genetics , Epigenomics/methods , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Promoter Regions, Genetic/genetics , RNA/genetics , Transcription Initiation Site
13.
J Cell Sci ; 126(Pt 10): 2256-66, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23525011

ABSTRACT

The miR-200 family is a key regulator of the epithelial-mesenchymal transition, however, its role in controlling the transition between cancer stem-cell-like and non-stem-cell-like phenotypes is not well understood. We utilized immortalized human mammary epithelial (HMLE) cells to investigate the regulation of the miR-200 family during their conversion to a stem-like phenotype. HMLE cells were found to be capable of spontaneous conversion from a non-stem to a stem-like phenotype and this conversion was accompanied by the loss of miR-200 expression. Stem-like cell fractions isolated from metastatic breast cancers also displayed loss of miR-200 indicating similar molecular changes may occur during breast cancer progression. The phenotypic change observed in HMLE cells was directly controlled by miR-200 because restoration of its expression decreased stem-like properties while promoting a transition to an epithelial phenotype. Investigation of the mechanisms controlling miR-200 expression revealed both DNA methylation and histone modifications were significantly altered in the stem-like and non-stem phenotypes. In particular, in the stem-like phenotype, the miR-200b-200a-429 cluster was silenced primarily through polycomb group-mediated histone modifications whereas the miR-200c-141 cluster was repressed by DNA methylation. These results indicate that the miR-200 family plays a crucial role in the transition between stem-like and non-stem phenotypes and that distinct epigenetic-based mechanisms regulate each miR-200 gene in this process. Therapy targeted against miR-200 family members and epigenetic modifications might therefore be applicable to breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Mammary Glands, Human/metabolism , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinogenesis/genetics , Cell Line, Transformed , DNA Methylation , Epigenetic Repression , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Histones/metabolism , Humans , Mammary Glands, Human/pathology , Molecular Targeted Therapy , Neoplasm Metastasis , Neoplastic Stem Cells/pathology , Promoter Regions, Genetic/genetics , Transgenes/genetics
14.
Can J Physiol Pharmacol ; 89(9): 617-22, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21846301

ABSTRACT

The aim of this study was to determine the morphology and position of the excitatory and inhibitory motor neurons to the human gastric sling and clasp fibers. Motor neurons were identified by retrograde staining with 1,1'-didodecyl 3,3,3',3'-indocarbocyanine perchlorate (DiI), and choline acetyltransferase (ChAT) or nitric oxide synthase (NOS) immunoreactivity was then determined in these motor neurons. In the sling preparations, 46% of the DiI-stained cells were aboral motor neurons, 43% were local motor neurons, and only 10% were descending motor neurons. Overall, 58% were immunoreactive for ChAT, and 36% for NOS (P = 0.042). Sixty-two percent of local, and 66% of aboral DiI-stained motor neurons were immunoreactive for ChAT. In the clasp preparations, 52% of the DiI-stained cells were descending motor neurons, 45% were local motor neurons, and only 3% were aboral neurons. Overall, 31% were immunoreactive for ChAT and 65% for NOS (P = 0.039). Eighty-five percent of the DiI-stained descending motor neurons were immunoreactive for NOS. All of the cells that were labeled adequately had a single axon and a number of filamentous or flattened lobular dendrites, and fitted into the broad category of Dogiel type I neurons. In conclusion, the majority of the motor neurons to the sling fibers were ChAT-positive excitatory neurons from the myenteric plexus of the stomach and the local region, and to the clasp were predominantly NOS-positive inhibitory neurons from the esophagus.


Subject(s)
Motor Neurons/physiology , Muscle, Smooth/cytology , Muscle, Smooth/innervation , Stomach/innervation , Carbocyanines/chemistry , Choline O-Acetyltransferase/metabolism , Esophagus/innervation , Esophagus/metabolism , Female , Fluorescent Dyes/chemistry , Gastric Mucosa/metabolism , Humans , Immunohistochemistry/methods , Male , Middle Aged , Motor Neurons/cytology , Motor Neurons/metabolism , Muscle, Smooth/metabolism , Myenteric Plexus/cytology , Myenteric Plexus/metabolism , Myenteric Plexus/physiology , Nitric Oxide Synthase/metabolism , Stomach/cytology
15.
PLoS One ; 6(7): e22513, 2011.
Article in English | MEDLINE | ID: mdl-21829465

ABSTRACT

Esophageal adenocarcinoma (EAC) has become a major concern in Western countries due to rapid rises in incidence coupled with very poor survival rates. One of the key risk factors for the development of this cancer is the presence of Barrett's esophagus (BE), which is believed to form in response to repeated gastro-esophageal reflux. In this study we performed comparative, genome-wide expression profiling (using Illumina whole-genome Beadarrays) on total RNA extracted from esophageal biopsy tissues from individuals with EAC, BE (in the absence of EAC) and those with normal squamous epithelium. We combined these data with publically accessible raw data from three similar studies to investigate key gene and ontology differences between these three tissue states. The results support the deduction that BE is a tissue with enhanced glycoprotein synthesis machinery (DPP4, ATP2A3, AGR2) designed to provide strong mucosal defenses aimed at resisting gastro-esophageal reflux. EAC exhibits the enhanced extracellular matrix remodeling (collagens, IGFBP7, PLAU) effects expected in an aggressive form of cancer, as well as evidence of reduced expression of genes associated with mucosal (MUC6, CA2, TFF1) and xenobiotic (AKR1C2, AKR1B10) defenses. When our results are compared to previous whole-genome expression profiling studies keratin, mucin, annexin and trefoil factor gene groups are the most frequently represented differentially expressed gene families. Eleven genes identified here are also represented in at least 3 other profiling studies. We used these genes to discriminate between squamous epithelium, BE and EAC within the two largest cohorts using a support vector machine leave one out cross validation (LOOCV) analysis. While this method was satisfactory for discriminating squamous epithelium and BE, it demonstrates the need for more detailed investigations into profiling changes between BE and EAC.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/genetics , Biomarkers, Tumor/genetics , Esophageal Neoplasms/genetics , Esophagus/metabolism , Gene Expression Profiling , Mucous Membrane/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Barrett Esophagus/metabolism , Barrett Esophagus/pathology , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Case-Control Studies , Cell Proliferation , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophagus/pathology , Female , Genome, Human , Humans , Male , Middle Aged , Mucous Membrane/pathology , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Support Vector Machine , Young Adult
16.
Mol Biol Cell ; 22(10): 1686-98, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21411626

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a form of cellular plasticity that is critical for embryonic development and tumor metastasis. A double-negative feedback loop involving the miR-200 family and ZEB (zinc finger E-box-binding homeobox) transcription factors has been postulated to control the balance between epithelial and mesenchymal states. Here we demonstrate using the epithelial Madin Darby canine kidney cell line model that, although manipulation of the ZEB/miR-200 balance is able to repeatedly switch cells between epithelial and mesenchymal states, the induction and maintenance of a stable mesenchymal phenotype requires the establishment of autocrine transforming growth factor-ß (TGF-ß) signaling to drive sustained ZEB expression. Furthermore, we show that prolonged autocrine TGF-ß signaling induced reversible DNA methylation of the miR-200 loci with corresponding changes in miR-200 levels. Collectively, these findings demonstrate the existence of an autocrine TGF-ß/ZEB/miR-200 signaling network that regulates plasticity between epithelial and mesenchymal states. We find a strong correlation between ZEBs and TGF-ß and negative correlations between miR-200 and TGF-ß and between miR-200 and ZEBs, in invasive ductal carcinomas, consistent with an autocrine TGF-ß/ZEB/miR-200 signaling network being active in breast cancers.


Subject(s)
Autocrine Communication , Epithelial-Mesenchymal Transition/genetics , Homeodomain Proteins/metabolism , MicroRNAs/genetics , Repressor Proteins/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Cell Line , Cofilin 2 , DNA Methylation , Dogs , Feedback, Physiological , Female , Homeodomain Proteins/genetics , Humans , MicroRNAs/metabolism , Repressor Proteins/genetics , Signal Transduction , Transcription Factors/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology , Up-Regulation , Zinc Finger E-box Binding Homeobox 2 , Zinc Finger E-box-Binding Homeobox 1
17.
Toxicol Sci ; 121(1): 110-22, 2011 May.
Article in English | MEDLINE | ID: mdl-21292642

ABSTRACT

Arsenic is a well-recognized human carcinogen, yet the mechanism by which it causes human cancer has not been elucidated. MicroRNAs (miRNAs) are a big family of small noncoding RNAs and negatively regulate the expression of a large number of protein-coding genes. We investigated the role of miRNAs in arsenic-induced human bronchial epithelial cell malignant transformation and tumor formation. We found that prolonged exposure of immortalized p53-knocked down human bronchial epithelial cells (p53(low)HBECs) to low levels of arsenite (NaAsO2, 2.5 µM) caused malignant transformation that was accompanied by epithelial to mesenchymal transition (EMT) and reduction in the levels of miR-200 family members. Stably reexpressing miR-200b in arsenite-transformed cells (As-p53(low)HBECs) completely reversed their transformed phenotypes, as evidenced by inhibition of colony formation in soft agar and prevention of xenograft tumor formation in nude mice. Moreover, stably expressing miR-200b alone in parental nontransformed p53(low)HBECs was sufficient to completely prevent arsenite exposure from inducing EMT and malignant transformation. Further mechanistic studies showed that depletion of miR-200 in arsenite-transformed cells involved induction of the EMT-inducing transcription factors zinc-finger E-box-binding homeobox factor 1 (ZEB1) and ZEB2 and increased methylation of miR-200 promoters. Stably expressing ZEB1 alone in parental nontransformed p53(low)HBECs was sufficient to deplete miR-200, induce EMT and cause cell transformation, phenocopying the oncogenic effect of 16-week arsenite exposure. These findings establish for the first time a causal role for depletion of miR-200b expression in human cell malignant transformation and tumor formation resulting from arsenic exposure.


Subject(s)
Arsenic/toxicity , Bronchi/drug effects , Cell Transformation, Neoplastic , MicroRNAs/physiology , Animals , Base Sequence , Bronchi/pathology , DNA Methylation , DNA Primers , Epithelial Cells/drug effects , Epithelial Cells/pathology , Humans , Mice , Mice, Nude , Reverse Transcriptase Polymerase Chain Reaction
18.
Biol Cell ; 102(12): 635-44, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20843300

ABSTRACT

BACKGROUND INFORMATION: Carcinoma of the oesophagus is the sixth leading cause of cancer death in the western world and is associated with a 5-year survival of less than 15%. Recent evidence suggests that stromal-epithelial interactions are fundamental in carcinogenesis. The advent of co-culture techniques permits the investigation of cross-talk between the stroma and epithelium in a physiological setting. We have characterized a histologically representative oesophageal organotypic model and have used it to compare the most commonly used squamous oesophageal cell line, HET-1A, with primary oesophageal squamous cells for use in studies of the oesophageal epithelium in vitro. RESULTS: When grown in an organotypic culture with normal fibroblasts, the oesophageal carcinoma cell lines OE21 (squamous) and OE19 (adenocarcinoma) morphologically resembled the tumour of origin with evidence of stromal invasion and mucus production, respectively. However, HET-1A cells, which were derived from normal squamous oesophageal cells, appeared dysplastic and failed to display evidence of squamous differentiation. By comparison with primary oesophageal epithelial cells, the HET-1A cells were highly proliferative and did not express the epithelial markers E-cadherin or CK5/6 (casein kinase 5/6), or the stratified epithelial marker ΔNp63, but did express the mesenchymal markers vimentin and N-cadherin. CONCLUSION: Studies of epithelial carcinogenesis will benefit from culture systems which allow manipulation of the stromal and epithelial layers independently. We have developed an organotypic culture using primary oesophageal squamous cells and fibroblasts in which a stratified epithelium with a proliferative basal layer that stains strongly for ΔNp63 develops. This model will be suitable for the study of the molecular events in the development of Barrett's oesophagus. The most commonly used normal oesophageal squamous cell line, HET-1A, does not have the characteristics of normal oesophageal squamous cells and should not be used in models of the normal oesophageal epithelium. Until more representative cell lines are available, future studies in oesophageal cancer will be reliant on the availability and manipulation of primary tissue.


Subject(s)
Barrett Esophagus/pathology , Carcinoma, Squamous Cell/pathology , Epithelial Cells/pathology , Esophageal Neoplasms/pathology , Adenocarcinoma/pathology , Antigens, CD/biosynthesis , Cadherins/biosynthesis , Casein Kinases/biosynthesis , Coculture Techniques , Epithelial Cells/metabolism , Esophagus/cytology , Humans , Membrane Proteins/biosynthesis , Vimentin/biosynthesis
19.
Cancer Epidemiol Biomarkers Prev ; 19(8): 2080-7, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20696664

ABSTRACT

BACKGROUND: Although most cases of esophageal squamous cell carcinoma (ESCC) in western populations have been attributed to high levels of exposure to tobacco and alcohol, infectious agents have been postulated as possible causes, particularly human papillomavirus (HPV). METHODS: To explore this issue, we analyzed HPV DNA prevalence and HPV types together with lifestyle factors, in relation to tumor stage and survival in a low-incidence population. Archived tumor samples from a nationwide cohort of 222 ESCC patients were tested for the presence of HPV DNA by PCR; positive samples were sequenced to determine HPV type, and p16(INK4a) status was assessed by immunohistochemistry. RESULTS: Of 222 ESCC patients, 8 tested HPV positive (prevalence, 3.6%; 95% confidence interval, 1.1-6.1%), of which 6 were HPV-16 positive and 2 were HPV-35 positive. Four of the eight HPV-positive tumors overexpressed p16(INK4a). None of 55 normal esophageal tissue samples from healthy participants had any detectable HPV. Although the numbers were low, it seemed that patients with HPV-positive ESCC tumors were younger than those with HPV-negative tumors (mean age, 60.8 versus 65.3 years, P = 0.18) and had higher body mass index (BMI) throughout life (mean current BMI of 25.1 for HPV positive, 22.2 for HPV negative, P = 0.08; mean BMI at 20 years of 25.8 for HPV positive, 22.1 for HPV negative, P = 0.003). We found no difference between patients with HPV-positive and HPV-negative tumors with respect to other lifestyle factors. CONCLUSIONS: These findings suggest a very low prevalence of HPV DNA in human ESCC. IMPACT: HPV is very unlikely to be a common cause of ESCC in Australia.


Subject(s)
Carcinoma, Squamous Cell/virology , DNA, Viral/analysis , Esophageal Neoplasms/virology , Papillomaviridae/classification , Papillomavirus Infections/complications , Adult , Age Factors , Aged , Australia/epidemiology , Body Mass Index , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Case-Control Studies , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Female , Humans , Life Style , Male , Middle Aged , Neoplasm Staging , Risk Factors , Survival Analysis
20.
Ann Surg ; 252(1): 63-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20562606

ABSTRACT

OBJECTIVE: We investigated the relationship between reflux and aberrant deoxyribonucleic acid (DNA) methylation, comparing methylation in the columnar epithelium following successful fundoplication to that in subjects with a failed fundoplication. SUMMARY BACKGROUND DATA: Gastroesophageal reflux is the main risk factor for Barrett esophagus and adenocarcinoma. In these diseases, there is a high level of DNA methylation. METHODS: We enrolled 41 patients with Barrett esophagus and a fundoplication at least 5 years earlier for a 24-hour pH study, endoscopy, and collection of biopsies. Biopsies were obtained from 17 Barrett esophagus subjects who had not undergone esophageal surgery. RESULTS: At the time of the study, 31 subjects were pH normal, 10 abnormal. Columnar biopsies were collected from 21 of the pH normal and 9 pH abnormal subjects, and all no surgery subjects. Complete regression of columnar mucosa was seen in 7 subjects with pH normal and 1 with pH abnormal. The length of Barrett esophagus did not differ between groups preoperatively, but was significantly less at the time of the study in the pH normal compared with pH abnormal or no surgery groups. Significantly, fewer genes were methylated in the pH normal than the pH abnormal or no surgery groups, which did not differ from each other. The number of methylated genes correlated with increased reflux, intestinal metaplasia, and increased columnar-lined esophagus length, but not acid-suppression medication. CONCLUSIONS: Fundoplication that reduces reflux to normal levels can lead to regression of the columnar mucosa. Reflux is associated with aberrant DNA methylation, and control of reflux reduces deleterious genomic changes associated with cancer.


Subject(s)
Barrett Esophagus/metabolism , Barrett Esophagus/surgery , DNA Methylation/physiology , Fundoplication , Gastroesophageal Reflux/prevention & control , Barrett Esophagus/pathology , Biopsy , Esophagus/pathology , Female , Gastroesophageal Reflux/pathology , Humans , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...