Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Transl Oncol ; 34: 101698, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37307644

ABSTRACT

Prostate cancer (PCa) is one of the leading causes of cancer-related deaths in men. Localised PCa can be treated effectively, but most patients relapse/progress to more aggressive disease. One possible mechanism underlying this progression is alternative splicing of the androgen receptor, with AR variant 7(ARV7) considered to play a major role. Using viability assays, we confirmed that ARV7-positive PCa cells were less sensitive to treatment with cabazitaxel and an anti-androgen-enzalutamide. Also, using live-holographic imaging, we showed that PCa cells with ARV7 exhibited an increased rate of cell division, proliferation, and motility, which could potentially contribute to a more aggressive phenotype. Furthermore, protein analysis demonstrated that ARV7 knock-down was associated with a decrease in insulin-like growth factor-2 (IGFBP-2) and forkhead box protein A1(FOXA1). This correlation was confirmed in-vivo using PCa tissue samples. Spearman rank correlation analysis showed significant positive associations between ARV7 and IGFBP-2 or FOXA1 in tissue from patients with PCa. This association was not present with the AR. These data suggest an interplay of FOXA1 and IGFBP-2 with ARV7-mediated acquisition of an aggressive prostate cancer phenotype.

2.
Cancer Lett ; 419: 187-202, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29331414

ABSTRACT

Since disturbed metabolic conditions such as obesity and diabetes can be critical determinants of breast cancer progression and therapeutic failure, we aimed to determine the mechanism responsible for their pro-oncogenic effects. Using non-invasive, epithelial-like ERα-positive MCF-7 and T47D human breast cancer cells we found that hyperglycaemia induced epithelial to mesenchymal transition (EMT), a key programme responsible for the development of metastatic disease. This was demonstrated by loss of the epithelial marker E-cadherin together with increases in mesenchymal markers such as vimentin, fibronectin and the transcription factor SLUG, together with an enhancement of cell growth and invasion. These phenotypic changes were only observed with cells grown on fibronectin and not with those plated on collagen. Analyzing metabolic parameters, we found that hyperglycaemia-induced, matrix-specific EMT promoted the Warburg effect by upregulating glucose uptake, lactate release and specific glycolytic enzymes and transporters. We showed that silencing of fatty acid synthase (FASN) and the downstream ERα, which we showed previously to mediate hyperglycaemia-induced chemoresistance in these cells, resulted in suppression of cell growth: however, this also resulted in a dramatic enhancement of cell invasion and SLUG mRNA levels via a novel caveolin-1-dependent mechanism.


Subject(s)
Caveolin 1/metabolism , Epithelial-Mesenchymal Transition/drug effects , Estrogen Receptor alpha/metabolism , Fatty Acid Synthase, Type I/metabolism , Glucose/pharmacology , Signal Transduction/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caveolin 1/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Estrogen Receptor alpha/genetics , Extracellular Matrix/metabolism , Fatty Acid Synthase, Type I/genetics , Female , Humans , Hyperglycemia/physiopathology , MCF-7 Cells , Neoplasm Invasiveness , RNA Interference , Signal Transduction/genetics
3.
Endocr Relat Cancer ; 24(1): 17-30, 2017 01.
Article in English | MEDLINE | ID: mdl-27754854

ABSTRACT

The incidence of many common cancers varies between different populations and appears to be affected by a Western lifestyle. Highly proliferative malignant cells require sufficient levels of nutrients for their anabolic activity. Therefore, targeting genes and pathways involved in metabolic pathways could yield future therapeutics. A common pathway implicated in energetic and nutritional requirements of a cell is the LKB1/AMPK pathway. Metformin is a widely studied anti-diabetic drug, which improves glycaemia in patients with type 2 diabetes by targeting this pathway. We investigated the effect of metformin on prostate cancer cell lines and evaluated its mechanism of action using DU145, LNCaP, PC3 and VCaP prostate cancer cell lines. Trypan blue dye-exclusion assay was used to assess levels of cell death. Western immunoblotting was used to determine the abundance of proteins. Insulin-like growth factor-binding protein-2 (IGFBP-2) and AMPK genes were silenced using siRNA. Effects on cell morphology were visualised using microscopy. IGFBP-2 gene expression was assessed using real-time RT-PCR. With DU145 and LNCaP cells metformin alone induced cell death, but this was reduced in hyperglycaemic conditions. Hyperglycaemia also reduced the sensitivity to Docetaxel, but this was countered by co-treatment with metformin. LKB1 was required for the activation of AMPK but was not essential to mediate the induction of cell death. An alternative pathway by which metformin exerted its action was through downregulation of IGFBP-2 in DU145 and LNCaP cells, independently of AMPK. This finding could have important implications in relation to therapeutic strategies in prostate cancer patients presenting with diabetes.


Subject(s)
Antineoplastic Agents/pharmacology , Hyperglycemia , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Prostatic Neoplasms/drug therapy , Taxoids/pharmacology , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Cell Death/drug effects , Cell Line, Tumor , Docetaxel , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Humans , Hyperglycemia/drug therapy , Hyperglycemia/genetics , Hyperglycemia/metabolism , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor Binding Protein 2/metabolism , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics
4.
Endocr Relat Cancer ; 23(2): 125-34, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26647383

ABSTRACT

Breast cancer patients with diabetes respond less well to chemotherapy; in keeping with this we determined previously that hyperglycaemia-induced chemoresistance in estrogen receptor (ERα) positive breast cancer cells and showed that this was mediated by fatty acid synthase (FASN). More recent evidence suggests that the effect of metabolic syndrome and diabetes is not the same for all subtypes of breast cancer with inferior disease-free survival and worse overall survival only found in women with ERα positive breast cancer and not for other subtypes. Here we examined the impact of hyperglycaemia on ERα negative breast cancer cells and further investigated the mechanism underlying chemoresistance in ERα with a view to identifying strategies to alleviate hyperglycaemia-induced chemoresistance. We found that hyperglycaemia-induced chemoresistance was only observed in ERα breast cancer cells and was dependent upon the expression of ERα as chemoresistance was negated when the ERα was silenced. Hyperglycaemia-induced an increase in activation and nuclear localisation of the ERα that was downstream of FASN and dependent on the activation of MAPK. We found that fulvestrant successfully negated the hyperglycaemia-induced chemoresistance, whereas tamoxifen had no effect. In summary our data suggests that the ERα may be a predictive marker of poor response to chemotherapy in breast cancer patients with diabetes. It further indicates that anti-estrogens could be an effective adjuvant to chemotherapy in such patients and indicates the importance for the personalised management of breast cancer patients with diabetes highlighting the need for clinical trials of tailored chemotherapy for diabetic patients diagnosed with ERα positive breast cancers.


Subject(s)
Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/physiology , Estrogen Receptor alpha/metabolism , Hyperglycemia/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Ceramides/pharmacology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hyperglycemia/pathology , Paclitaxel/pharmacology
5.
Endocr Relat Cancer ; 20(5): 741-51, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23959956

ABSTRACT

Clinically relevant prostate cancer (PCa) is more frequent in Westernised societies and increasingly men have co-morbidities associated with a Western lifestyle, primarily diabetes, characterised by hyperinsulinaemia and hyperglycaemia. IGFs and their binding proteins (IGFBPs) are important mediators of the effects of nutrition on growth and play a key role in the development of PCa. We used DU145, PC3 and LNCaP PCa cell lines to examine how hyperglycaemia altered their response to docetaxel. Trypan Blue dye-exclusion assay was used to determine the percentage of cell death. Protein abundance was determined using western immunoblotting. Levels of IGFBP2 were measured using an ELISA. IGFBP2 gene silencing was achieved using siRNA technology. DNA methylation was assessed using combined bisulphide restriction analysis. Acetylation status of histones H3 and H4 associated with IGFBP2 gene was assessed using chromatin immunoprecipitation assay. Hyperglycaemia reduced docetaxel-induced apoptosis by 40% for DU145 cells and by 88% for LNCaP cells. This reduced cell death was mediated by a glucose-induced up-regulation of IGFBP2, as silencing IGFBP2 negated the survival effect of high glucose. Glucose increased IGFBP2 via increasing the acetylation of histones associated with the IGFBP2 gene promoter. This finding could have important implications in relation to therapeutic strategies as epigenetic modulation could be reversible.


Subject(s)
Drug Resistance, Neoplasm/physiology , Hyperglycemia/metabolism , Insulin-Like Growth Factor Binding Protein 2/metabolism , Prostatic Neoplasms/metabolism , Acetylation , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Cell Line, Tumor , Docetaxel , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Insulin-Like Growth Factor Binding Protein 2/genetics , Male , Naphthols/pharmacology , Promoter Regions, Genetic , Prostatic Neoplasms/drug therapy , RNA, Small Interfering/genetics , Sirtuin 1/antagonists & inhibitors , Sirtuin 2/antagonists & inhibitors , Taxoids/pharmacology
6.
J Pathol ; 230(1): 95-106, 2013 May.
Article in English | MEDLINE | ID: mdl-23299523

ABSTRACT

Podocytes are crucial for preventing the passage of albumin into the urine and, when lost, are associated with the development of albuminuria, renal failure and cardiovascular disease. Podocytes have limited capacity to regenerate, therefore pro-survival mechanisms are critically important. Insulin-like growth factor-II (IGF-II) is a potent survival and growth factor; however, its major function is thought to be in prenatal development, when circulating levels are high. IGF-II has only previously been reported to continue to be expressed in discrete regions of the brain into adulthood in rodents, with systemic levels being undetectable. Using conditionally immortalized human and ex vivo adult mouse cells of the glomerulus, we demonstrated the podocyte to be the major glomerular source and target of IGF-II; it signals to this cell via the IGF-I receptor via the PI3 kinase and MAPK pathways. Functionally, a reduction in IGF signalling causes podocyte cell death in vitro and glomerular disease in vivo in an aged IGF-II transgenic mouse that produces approximately 60% of IGF-II due to a lack of the P2 promoter of this gene. Collectively, this work reveals the fundamental importance of IGF-II in the mature podocyte for glomerular health across mammalian species.


Subject(s)
Insulin-Like Growth Factor II/metabolism , Podocytes/cytology , Podocytes/metabolism , Signal Transduction/physiology , Aging/physiology , Animals , Cell Line, Transformed , Cell Survival/physiology , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/genetics , Kidney Glomerulus/cytology , Kidney Glomerulus/physiology , Mesangial Cells/cytology , Mesangial Cells/metabolism , Mice , Mice, Transgenic , RNA, Small Interfering/genetics
7.
Minerva Endocrinol ; 37(2): 173-85, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22691890

ABSTRACT

The major issue currently being faced in the management of prostate cancer is the inability to distinguish between indolent prostate tumors that will not present clinically from more aggressive and metastatic prostate cancers that will impact on men's lives. Only a small proportion of prostate cancers can be accounted for by unmistakable hereditary cancer syndromes and the predominant contribution to the progression of most sporadic cancers is thought to be environmental, with nutrition having the greatest influence. Population studies have clearly implicated metabolic factors as contributors to disease progression and poor response to therapy. It is well established that the IGF system is key in regulating growth and metabolism and mediates the effects of nutrition on these processes. It consists of two ligands (IGF-I and IGF-II), two receptors [type 1 IGF-IR and IGF-II/mannose 6-phosphate receptor], and six high affinity IGF-binding proteins (IGFBP-1 to -6). This review provides evidence from in vitro, in vivo, clinical and epidemiology studies that indicates an important role for the IGF axis in the development of prostate cancer and the likely role that it plays in mediating the effects of nutrition on disease progression. We suggest that the IGF axis is central to understanding how lifestyle impacts on prostate cancer and we highlight this by describing numerous strategies being developed to target this axis.


Subject(s)
Adenocarcinoma/metabolism , Neoplasm Proteins/physiology , Prostatic Neoplasms/metabolism , Receptors, Somatomedin/physiology , Somatomedins/physiology , Adenocarcinoma/epidemiology , Adenocarcinoma/physiopathology , Adenocarcinoma/therapy , Androgens/metabolism , Antineoplastic Agents, Hormonal/therapeutic use , Disease Progression , Energy Metabolism , Humans , Incidence , Insulin-Like Growth Factor Binding Proteins , Male , Molecular Targeted Therapy , Neoplasms, Hormone-Dependent/epidemiology , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/physiopathology , Neoplasms, Hormone-Dependent/therapy , Overnutrition/complications , Overnutrition/metabolism , Overnutrition/physiopathology , Prevalence , Prostatectomy , Prostatic Neoplasms/epidemiology , Prostatic Neoplasms/physiopathology , Prostatic Neoplasms/therapy , Tumor Cells, Cultured , Zinc/metabolism
8.
Oncogene ; 31(13): 1710-22, 2012 Mar 29.
Article in English | MEDLINE | ID: mdl-21892205

ABSTRACT

Merlin is a tumour suppressor involved in the development of a variety of tumours including mesotheliomas. Neurofibromatosis type 2 (NF2), a dominantly inherited tumour disease, is also caused by loss of merlin. NF2 patients suffer from multiple genetically well-defined tumours, schwannomas are most frequent among those. Using our in vitro model for human schwannoma, we found that schwannoma cells display enhanced proliferation because of the overexpression/activation of platelet-derived growth factor receptor and ErbB2/3, increased cell-matrix adhesion because of the overexpression of integrins, and decreased apoptosis. Mechanisms underlying schwannomas basal proliferation and cell-matrix adhesion are not understood. Here, we investigated insulin-like growth factor-binding protein-1 (IGFBP-1), which is expressed and released from central nervous system tumours and strongly overexpressed in schwannoma at the mRNA level. IGFBP-1 acts via ß1-integrin and focal-adhesion-kinase (FAK), which are strongly overexpressed and basally activated in schwannoma. Using short hairpin RNA knockdown, small inhibitors and recombinant IGFBP-1, we demonstrate that schwannoma cells, in contrast to Schwann cells, release IGFBP-1 that activates the Src/FAK pathway, via integrin ß1, potentiating schwannoma's proliferation and cell-matrix adhesion. We show that FAK localizes to the nucleus and Src triggers IGFBP-1 production. Further, we observed downregulation of the tumour-suppressor phosphatase and tensin homolog in schwannoma cells leading to increased activity of anti-apoptotic AKT. Thus, IGFBP-1/integrin ß1/Src/FAK pathway has a crucial role in merlin-related tumourigenesis and therefore represents an important therapeutic target in the treatment of merlin-deficient tumours.


Subject(s)
Insulin-Like Growth Factor Binding Protein 1/physiology , Neurilemmoma/metabolism , Cell Adhesion , Cell Proliferation , Cell Survival , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 1/metabolism , Membrane Proteins/metabolism , Oncogene Protein v-akt/metabolism , PTEN Phosphohydrolase/metabolism , Schwann Cells/metabolism , Signal Transduction/physiology , src-Family Kinases/metabolism
9.
Br J Cancer ; 104(10): 1587-93, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21487405

ABSTRACT

BACKGROUND: The development of androgen independence, chemo-, and radioresistance are critical markers of prostate cancer progression and the predominant reasons for its high mortality. Understanding the resistance to therapy could aid the development of more effective treatments. AIM: The aim of this study is to investigate the effects of insulin-like growth factor-binding protein-2 (IGFBP-2) on prostate cancer cell proliferation and its effects on the response to docetaxel. METHODS: DU145 and PC3 cells were treated with IGFBP-2, insulin-like growth factor I (IGF-I) alone or in combination with blockade of the IGF-I receptor or integrin receptors. Cells were also treated with IGFBP-2 short interfering ribonucleic acid with or without a PTEN (phosphatase and tensin homologue deleted on chromosome 10) inhibitor or docetaxel. Tritiated thymidine incorporation was used to measure cell proliferation and Trypan blue cell counting for cell death. Levels of IGFBP-2 mRNA were measured using RT-PCR. Abundance and phosphorylation of proteins were assessed using western immunoblotting. RESULTS: The IGFBP-2 promoted cell growth in both cell lines but with PC3 cells this was in an IGF-dependent manner, whereas with DU145 cells the effect was independent of IGF receptor activation. This IGF-independent effect of IGFBP-2 was mediated by interaction with ß-1-containing integrins and a consequent increase in PTEN phosphorylation. We also determined that silencing IGFBP-2 in both cell lines increased the sensitivity of the cells to docetaxel. CONCLUSION: The IGFBP-2 has a key role in the growth of prostate cancer cells, and silencing IGFBP-2 expression reduced the resistance of these cells to docetaxel. Targeting IGFBP-2 may increase the efficacy of docetaxel.


Subject(s)
Antineoplastic Agents/pharmacology , Insulin-Like Growth Factor Binding Protein 2/pharmacology , Insulin-Like Growth Factor I/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Taxoids/pharmacology , Cell Growth Processes/drug effects , Cell Line, Tumor , Docetaxel , Humans , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor Binding Protein 2/metabolism , Insulin-Like Growth Factor I/genetics , Male , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Recombinant Proteins/pharmacology , Transfection
10.
Endocr Relat Cancer ; 17(2): 539-51, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20356977

ABSTRACT

The prognosis for women with breast cancer is adversely affected by the comorbidities of obesity and diabetes mellitus (DM), which are conditions associated with elevated levels of circulating fatty acids, hyperglycaemia and hyperinsulinaemia. We investigated the effects of exposure of non-malignant and malignant human breast epithelial cells to elevated levels of fatty acids and glucose on their growth, survival and response to chemotherapeutic agents. We found that palmitate induced cell death in the non-malignant cells but not in the malignant cells, which was abrogated through the inhibition of ceramide production and by oleate but not by IGF1. Fatty acid synthase (FAS) is responsible for the de novo synthesis of fatty acids from sugars, and is over-expressed in many epithelial cancers. Abundance of FAS was higher in malignant cells than in non-malignant cells, and was up-regulated by IGF1 in both cell types. IGF-induced growth of non-malignant cells was unaffected by suppression of FAS expression, whereas that of malignant cells was blocked as was their resistance to palmitate-induced cell death. Palmitate did not affect cell proliferation, whereas oleate promoted the growth of non-malignant cells but had the opposite effect, that is, inhibition of IGF1-induced growth of malignant cells. However, when the phosphatidylinositol 3-kinase pathway was inhibited, oleate enhanced IGF1-induced growth in both cell types. Hyperglycaemia conferred resistance on malignant cells, but not on non-malignant cells, to chemotherapy-induced cell death. This resistance was overcome by inhibiting FAS or ceramide production. Understanding the mechanisms involved in the associations between obesity, DM and breast cancer may lead to more effective treatment regimens and new therapeutic targets.


Subject(s)
Breast Neoplasms/drug therapy , Carcinoma/drug therapy , Drug Resistance, Neoplasm , Fatty Acid Synthases/physiology , Hyperglycemia/complications , Antineoplastic Agents/therapeutic use , Breast Neoplasms/complications , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma/complications , Carcinoma/metabolism , Carcinoma/pathology , Cell Death/drug effects , Ceramides/adverse effects , Ceramides/metabolism , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , Fatty Acid Synthases/antagonists & inhibitors , Fatty Acid Synthases/metabolism , Fatty Acids/adverse effects , Fatty Acids/metabolism , Fatty Acids/pharmacology , Female , Humans , Hyperglycemia/metabolism , Hyperglycemia/pathology , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism , Mammary Glands, Human/physiology , Palmitic Acid/pharmacology , RNA, Small Interfering/pharmacology , Tumor Cells, Cultured
11.
Oncogene ; 26(40): 5966-72, 2007 Aug 30.
Article in English | MEDLINE | ID: mdl-17369847

ABSTRACT

The dual-function phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is the second most frequently mutated gene in human cancers. PTEN counteracts the functions of many growth factors, the most prevalent of which is insulin-like growth factor II (IGF-II). PTEN expression is stimulated by IGF-II forming a feedback loop. Investigating IGF-binding protein (IGFBP) modulation of IGF-II actions on MCF-7 breast cancer cells, we found that IGFBP-2 also regulates PTEN. The MCF-7 cells were not responsive to high doses of IGF-II due to induction of PTEN, which was not observed with an IGF-II-analog that does not bind to IGFBPs or in the presence of an inhibitor that prevents IGFs associating with IGFBPs. These cells predominantly produce IGFBP-2: blocking IGFBP-2 with a specific antibody, or preventing IGFBP-2 binding to integrins, restored the induction of PTEN and the cells were non-responsive to high doses of the IGF-II-analog. Our findings indicate that breast cancer cells do not respond to high doses of IGF-II due to induction of PTEN, but IGFBP-2, when free from IGF-II can suppress PTEN. Levels of IGFBP-2 are elevated frequently in human tumors: its ability to regulate PTEN could have important implications in relation to therapeutic strategies targeting growth factor pathways.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor Binding Protein 2/physiology , Insulin-Like Growth Factor II/physiology , PTEN Phosphohydrolase/biosynthesis , Cell Line, Tumor , Disease Progression , Dose-Response Relationship, Drug , Humans , Integrins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Oligopeptides/chemistry , Peptide Fragments/chemistry , Signal Transduction , Somatomedins/metabolism
12.
Endocrinology ; 147(7): 3484-500, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16614079

ABSTRACT

IGF-binding protein (IGFBP)-3 is generally considered to have actions that counterbalance those of IGFs and is therefore being developed as a cancer treatment. In breast tumors, however, high levels are associated with aggressive tumors and poor prognosis. Consistent with this we have demonstrated that although IGFBP-3 and a non-IGF-binding fragment (serine phosphorylation domain peptide) reduced attachment and enhanced apoptosis of Hs578T breast cancer cells cultured on collagen or laminin, it promoted their attachment and survival on fibronectin, which is abundant in the matrix of aggressive tumors. We have now examined the factors that determine whether IGFBP-3 has positive or negative actions on breast epithelial cells. IGFBP-3 also promoted survival of Hs578T cells in the presence of an antibody to the beta1-integrin subunit or when cholesterol-stabilized complexes were disrupted. These actions were blocked by IGF-I or a MAPK inhibitor. Serine phosphorylation domain peptide had similar actions on MCF-7 cells that were again reversed on fibronectin or with disruption of cholesterol-stabilized complexes and blocked by the beta1-integrin antibody. In contrast, IGFBP-3 promoted growth and survival for nonmalignant MCF-10A cells, but these effects were again reversed on fibronectin and blocked by the beta1 antibody or a MAPK inhibitor or by disruption of cholesterol-stabilized complexes. On Hs578T cells, IGFBP-3 bound to caveolin-1 and beta1-integrins, enhancing their aggregation, the recruitment of focal adhesion kinase, and the activation of MAPK. In summary, with three breast epithelial cell lines, IGFBP-3 had positive or negative effects on growth and survival dependent upon the status of cholesterol-stabilized integrin receptor complexes.


Subject(s)
Breast Neoplasms/pathology , Breast/pathology , Cholesterol/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor Binding Protein 3/physiology , Apoptosis , Caveolin 1/metabolism , Cell Line, Tumor , Detergents/pharmacology , Disease Progression , Fibronectins/metabolism , Humans , Integrin beta1/metabolism , Phosphorylation , Transferrin/metabolism
13.
Eur J Cancer ; 41(7): 1017-25, 2005 May.
Article in English | MEDLINE | ID: mdl-15862750

ABSTRACT

Activation of protein kinase-B/Akt (pAkt) is mediated by oestrogen and involves HER-2 in vitro, to phosphorylate Hdm2 and influence p53 cytoplasmic localisation and degradation. Expression of all active Akt isoforms (pAkt) were examined, together with p53/Hdm2 subcellular expression in invasive ductal breast cancers (IDCs), to evaluate whether in vitro findings were related to clinical data and determine the effect on outcome. Immunohistochemical expression of serine 473 specific phosphorylated Akt (pAkt) isoforms (Akt-1,2,3) was evaluated in 97 patients, together with subcellular expression of p53/Hdm2. The results show that pAkt was evaluable in 95 patients with cytoplasmic expression in 81% and more likely to be associated with larger tumours (P=0.007), with no correlation with HER-2 expression. pAkt correlated with increasing levels of cytoplasmic p53 (P=0.025) and was associated with a reduced disease-free survival (P=0.04; univariate). In conclusion, pAkt has implications in breast cancer growth through mechanisms inactivating p53 with an association with immunohistochemical p53 expression, which is preferentially cytoplasmic. Despite in vitro associations, pAkt appears to be a variable marker of HER-2 expression.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Intraductal, Noninfiltrating/metabolism , Gene Products, tat/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Adult , Aged , Aged, 80 and over , Female , Genetic Markers/genetics , Humans , Immunohistochemistry , Middle Aged , Prognosis , Proto-Oncogene Proteins c-akt , Receptor, ErbB-2/metabolism , Survival Analysis
14.
Breast Cancer Res ; 6(5): R571-85, 2004.
Article in English | MEDLINE | ID: mdl-15318938

ABSTRACT

INTRODUCTION: p14ARF stabilises nuclear p53, with a variable expression of p14ARF mRNA in breast cancers. In vitro, nuclear p14ARF binds Hdm2 to block Hdm2-dependent nucleocytoplasmic shuttling of p53, which is required before cytoplasmic degradation of p53. p14ARF is negatively regulated by p53 and through p53-independent pathways. No studies have yet examined levels of p14ARF protein expression in breast cancer and their relationship to Hdm2/p53 immunoreactivity or subcellular localisation. Previously, immunohistochemical expression of cytoplasmic p14ARF, p53 and Hdm2 has been described. HER-2 (c-erbB2/neu) predicts prognosis and interacts with the p14ARF/Hdm2 pathway to inactivate p14ARF and to influence Hdm2 activity and localisation. This study examined p14ARF and p53/Hdm2 expression and subcellular localisation by using immunohistochemistry in a series of invasive ductal breast cancers (IDCs) with concomitant ductal carcinoma in situ (DCIS), to evaluate whether findings in vitro were related to clinicopathological parameters such as HER-2 and their effect on patient outcome. METHODS: The 4C6 anti-p14ARF monoclonal antibody and Dako Envision Plus system were used to evaluate p14ARF expression in 103 patients; p53/Hdm2 staining was performed. RESULTS: p14ARF was evaluable in 96 patients, with nuclear p14ARF expression (modified Quick-score > or = 3) in 79% (n = 76) of IDCs and in associated DCIS in 74 patients. Cytoplasmic p14ARF was detectable in 23 breast cancers. Nuclear and cytoplasmic p14ARF showed no correlation with p53 subcellular immunoreactivity. Increasing levels of cytoplasmic p14ARF were associated with nuclear and cytoplasmic Hdm2 expression (P < 0.001). Subcellular ARF expression was not associated with clinicopathological parameters, and although not an independent prognosticator, these preliminary findings suggest that cytoplasmic p14ARF might be associated with a better overall survival (P = 0.09; log rank). The association between HER-2 positivity and nuclear p14ARF (P = 0.038), as well as nuclear Hdm2 (P = 0.019), reflects the in vitro findings of HER-2 interaction with the ARF/Hdm2 pathway. Cytoplasmic p53 and Hdm2 expression might have biological implications, through an association of cytoplasmic p53 with increased tumour proliferation (P = 0.005), and an improved overall survival (P = 0.002, log rank) in cytoplasmic Hdm2-expressing tumours, that independently predict favourable overall survival (P = 0.02) and disease-free survival (P = 0.03). CONCLUSIONS: Nuclear p14ARF expression is similar in IDCs and DCIS and is associated with Hdm2 immunoreactivity. Nuclear p14ARF and Hdm2 might be regulated by HER-2. Clearly, our findings in vivo suggest a complexity of p14ARF/Hdm2 and p53 pathways in which consideration of cytoplasmic p14ARF and Hdm2 might have tumorigenic implications.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Intraductal, Noninfiltrating/metabolism , Tumor Suppressor Protein p14ARF/metabolism , Adult , Aged , Aged, 80 and over , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/mortality , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/mortality , Carcinoma, Intraductal, Noninfiltrating/pathology , Humans , Immunohistochemistry , Middle Aged , Nuclear Proteins/metabolism , Prognosis , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Receptor, ErbB-2/metabolism , Survival Analysis , Tumor Suppressor Protein p53/metabolism
15.
Br J Cancer ; 91(2): 305-11, 2004 Jul 19.
Article in English | MEDLINE | ID: mdl-15213724

ABSTRACT

Human breast cancer is the leading cause of cancer death in women from Western societies, and a large study of the epidemiology demonstrated strong associations between human prolactin and risk of breast cancer. Using established models of apoptosis of human breast cancer cell lines, we assessed the role of prolactin in breast cancer cell growth and survival. We showed that prolactin had no effect on the metabolic activity or total cell number of any cell lines. We confirmed endogenous prolactin production by these cells and that the levels varied. In the presence of a prolactin-neutralising antibody, each of the cell lines responded with the induction of apoptosis as opposed to growth inhibition. The sensitivity of the cell lines to the physiological inducer of apoptosis, C2-ceramide, appeared relative to the levels of endogenous prolactin that they contained. We then showed that exogenously added prolactin acted as a potent survival factor against apoptosis in all the cell lines examined. In addition, we demonstrated that a prolactin-neutralising antibody in combination with C2-ceramide caused an anticipated, additive increase in cell death. This study demonstrated that prolactin protects human breast cancer cell lines against apoptosis and this may have important implications for cancer treatment.


Subject(s)
Breast Neoplasms/metabolism , Prolactin/physiology , Sphingosine/analogs & derivatives , Apoptosis/drug effects , Apoptosis/physiology , Breast Neoplasms/pathology , Cell Survival/physiology , Combined Modality Therapy , Enzyme Inhibitors/pharmacology , Female , Humans , Sphingosine/pharmacology , Tumor Cells, Cultured
16.
Hum Reprod ; 18(12): 2672-7, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14645190

ABSTRACT

BACKGROUND: The role of prolactin in the regulation of ovarian folliculogenesis and corpus luteal function and in particular its relationship to atresia in these structures is as yet unclear. We established a model of apoptosis in which to examine the actions of prolactin. METHOD: Granulosa cells collected from IVF-flush were cultured at 0.1-0.3 x 10(6) cells/well in growth media for 48 h, placed into serum-free media for 24 h prior to dosing for 24 h. Dose responses to C2-ceramide and prolactin were performed. Cells were then treated with an apoptotic dose of C2-ceramide alone, prolactin (100 ng/ml) alone or a combination of the two. Cell death was assessed by Trypan Blue cell counting and MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; Thiazolyl Blue] assay and apoptosis confirmed by morphological assessment and flow cytometry. RESULTS: C2-ceramide (0-40 micro mol/l) induced a dose-dependent increase in cell death (63.8% increase at 40 micro mol/l) and, morphologically, cells exhibited classical features of apoptosis. Prolactin alone had no effect on metabolic activity or total cell number. On co- incubation, prolactin alone had no effect on cell death, whereas C2-ceramide induced an approximately 62.6% increase in apoptosis, which was inhibited in the presence of prolactin. CONCLUSIONS: Prolactin may contribute significantly to early corpus luteum formation and survival by acting as a potent antiapoptotic factor for human granulosa cells.


Subject(s)
Apoptosis/drug effects , Granulosa Cells/cytology , Prolactin/administration & dosage , Sphingosine/analogs & derivatives , Sphingosine/administration & dosage , Cell Count , Cell Survival/drug effects , Cells, Cultured , Corpus Luteum/physiology , Dose-Response Relationship, Drug , Female , Flow Cytometry , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Humans , Progesterone/analysis , Progesterone/biosynthesis , Prolactin/analysis , Prolactin/physiology
17.
Exp Dermatol ; 12(4): 426-34, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12930299

ABSTRACT

In this study, we have examined the effects of retinoic acid (RA) on the human immortalized keratinocyte cell line (HaCaT). A significant twofold (P < 0.01) increase in apoptotic cell death compared with the control was found within 24 h of treatment with 10-5 M of RA. Apoptosis was confirmed by flow cytometry. Cycloheximide did not inhibit this acute RA-induced apoptosis. Interestingly, insulin-like growth factor-II (IGF-II, 50 ng/ml) was able to significantly (67.3%; P < 0.05) reduce RA effects, whereas IGF-I (50 ng/ml) and insulin (75 ng/ml) were without effect. Furthermore, analogues of IGF-II [leu27 IGF-II and Des(1-6) IGF-II], with altered affinities for the IGF-I receptor and IGF-binding proteins (IGFBPs), but retained affinities for the IGF-II receptor, also completely inhibited (100%; P < 0.01) RA-induced apoptosis, while an IGF-I receptor antagonist did not reduce the survival effects of IGF-II. Insulin pretreatment negates the survival effect of IGF-II. In contrast, mannose 6 phosphate (M6P) did not alter RA or IGF-II actions. These results indicate that rapid induction of cell death by RA is independent of production or secretion of new proteins. The inhibition of RA action by IGF-II was independent of its ability to signal through the IGF-I receptor or to interact with IGFBPs.


Subject(s)
Apoptosis/drug effects , Insulin-Like Growth Factor II/pharmacology , Keratinocytes/drug effects , Keratinocytes/metabolism , Receptor, IGF Type 2/metabolism , Tretinoin/pharmacology , Cell Division/drug effects , Cell Line , Cycloheximide/pharmacology , Humans , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Keratinocytes/cytology , Mannosephosphates/pharmacology , Receptor, IGF Type 1/drug effects , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 2/drug effects , Recombinant Proteins/pharmacology
18.
Carcinogenesis ; 24(3): 393-401, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12663497

ABSTRACT

Sodium butyrate (NaBt) and the pro-apoptotic IGFBP-3 protein, expressed at the top of the normal colonic crypt, have both been implicated in the regulation of apoptosis in colonic epithelial cells. Recent studies in human breast and hepatic cell lines have shown that NaBt can transcriptionally upregulate IGFBP-3 expression. However, the role of butyrate in the regulation of IGFBP-3 expression in the colon is less clear, with reports of both up- and downregulation of the IGFBP-3 protein in colorectal cancer cell lines. In this study we have shown that the level of IGFBP-3 protein expression in colonic epithelial cells correlates with the p53 status of the cells; wildtype p53 cells secrete higher levels of IGFBP3 protein than mutant p53 cell lines. Data presented shows that, when treated with a dose of NaBt that induced significant apoptosis (4 mM for 48 h), there was an upregulation of IGFBP-3 protein in both wildtype and mutant p53 expressing cell lines. The NaBt-induced increase in secreted IGFBP-3 protein was associated with transcriptional upregulation of the IGFBP-3 gene. Using a transfected derivative of the S/RG/C2 adenoma-derived cell line, which stably expressed exogenous IGFBP-3 protein at levels equivalent to that secreted by the 4 mM NaBt-treated parental line (1-3 ng/10(6) cells), we have shown a >2-fold increase in the sensitivity of the cells to NaBt-induced apoptosis when compared with the vector control and parental cell lines. Furthermore, inhibition of the secreted IGFBP-3 protein, by addition of neutralizing antibodies, resulted in a significant decrease in NaBt-induced apoptosis. These data suggest that IGFBP-3 may act as a positive regulator of NaBt-induced apoptosis in colonic epithelial cells, and represents a potentially important mechanism whereby the sensitivity of colonic epithelial cells to NaBt-induced apoptosis can be increased.


Subject(s)
Adenoma/pathology , Apoptosis/physiology , Butyric Acid/pharmacology , Colonic Neoplasms/pathology , Insulin-Like Growth Factor Binding Protein 3/genetics , Somatomedins/physiology , Transcription, Genetic/drug effects , Up-Regulation/drug effects , Blotting, Northern , Epithelial Cells/cytology , Humans , Tumor Cells, Cultured
19.
J Cell Biochem ; 86(3): 583-9, 2002.
Article in English | MEDLINE | ID: mdl-12210764

ABSTRACT

Insulin-like growth factor binding protein-3 (IGFBP-3) is one of six high affinity-binding proteins that share a common function in regulating the bioavailability of the insulin-like growth factors. The six binding proteins have highly conserved C- and N-terminals that are essential to this function. Additionally, they all have specific functions on cellular homeostasis independent to the regulation of the insulin-like growth factors. It has previously been shown that insulin-like growth factor binding protein-3 can accentuate UV-induced apoptosis in a human carcinoma cell line. Using the KYSE 190 oesophageal carcinoma cell line we have demonstrated that a 15 amino acid (aa) peptide that lies within the mid-region of the protein can mimic the effect of the intact protein. This region contains the serine residues Ser(111) and Ser(113). Using two protocols, we modified these serine residues and have shown that both phosphorylation and derivatization of IGFBP-3 can negate the accentuation of UV-induced cell death. These three independent pieces of evidence support the hypothesis that the variable mid-region is responsible for the specific pro-apoptotic functions of IGFBP-3, and suggest that phosphorylation may provide a mechanism for regulation of this action.


Subject(s)
Insulin-Like Growth Factor Binding Protein 3/chemistry , Insulin-Like Growth Factor Binding Protein 3/metabolism , Somatomedins/physiology , Amino Acid Sequence , Apoptosis/drug effects , Apoptosis/radiation effects , Creatine Kinase/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Phosphorylation , Structure-Activity Relationship , Tumor Cells, Cultured , Ultraviolet Rays
20.
Biochem Biophys Res Commun ; 294(5): 988-94, 2002 Jun 28.
Article in English | MEDLINE | ID: mdl-12074574

ABSTRACT

We demonstrated previously that IGFBP-3 alone had no effect on cell death, but dramatically modulated apoptosis in Hs578T IGF non-responsive cells. We investigated whether a non-IGF binding mutant of IGFBP-3 retained its intrinsic actions in this cell line, prior to investigating its actions in IGF-responsive cells (MCF-7 and MCF-10A). In the Hs578T cells, the ceramide analogue, C2-induced apoptosis, non-glycosylated, glycosylated or mutant IGFBP-3 alone had no effect but on co-incubation with C2, all forms of IGFBP-3 markedly accentuated triggered apoptosis. In MCF-7 cells, IGFBP-3 was unable to modulate C2-induced death. In the MCF-10A cells, IGFBP-3 acted as a potent survival factor. IGFBP-3 also affected cell growth in the MCF-10A cells (inhibiting at low doses but increasing growth at higher concentrations). These actions of IGFBP-3 in the MCF-10A cells were independent of IGF-1. IGFBP-3 has differential IGF-independent effects on cell death and growth in normal breast and breast cancer cells.


Subject(s)
Breast/cytology , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Sphingosine/analogs & derivatives , Apoptosis , Cell Division/drug effects , Cell Line , Dose-Response Relationship, Drug , Epithelial Cells/cytology , Epithelial Cells/drug effects , Female , Humans , Insulin-Like Growth Factor I/pharmacology , Mutation , Somatomedins/metabolism , Sphingosine/antagonists & inhibitors , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...