Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Nucl Med Biol ; 42(3): 274-82, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25542669

ABSTRACT

INTRODUCTION: Organic Anion Transporting Polypeptides (OATP) are a family of membrane associated transporters that facilitate estrone-3-sulphate (E3S) uptake by hormone dependent, post-menopausal breast cancers. We have established E3S as a potential ligand for targeting hormone dependent breast cancer cells, and in this study sought to prepare and investigate radioiodinated E3S as a tool to study the OATP system. METHODS: 2- and 4-Iodoestrone-3-sulfates were prepared from estrone via aromatic iodination followed by a rapid and high yielding sulfation procedure. The resulting isomers were separated by preparative HPLC and verified by (1)H NMR and analytical HPLC. Transport studies of 2- and 4-[(125)I]-E3S were conducted in hormone dependent (i.e. MCF-7) and hormone independent (i.e. MDA-MB-231) breast cancer cells in the presence or absence of the specific transport inhibitor, bromosulfophthalein (BSP). Cellular localization of OATP1A2, OATP2B1, OATP3A1 and OATP4A1 were determined by immunofluorescence analysis using anti-Na(+)/K(+) ATPase-α (1:100 dilution) and DAPI as plasma membrane and nuclear markers, respectively. RESULTS: Significantly (p<0.01) higher total accumulation of 2-[(125)I]-E3S was observed in hormone dependent MCF-7 as compared to hormone independent MDA-MB-231 breast cancer cells. In contrast 4-[(125)I]-E3S did not show cellular accumulation in either case. The efficiency of 2-[(125)I]-E3S transport (expressed as a ratio of Vmax/Km) was 2.4 times greater in the MCF-7 as compared to the MDA-MB-231 breast cancer cells. OATP1A2, OATP3A1 and OATP4A1 expression was localized in plasma membranes of MCF-7 and MDA-MB-231 cells confirming the functional role of these transporters in radioiodinated E3S cellular uptake. CONCLUSION: An efficient method for the preparation of 2- and 4-[(125)I]-E3S was developed and where the former demonstrated potential as an in vitro probe for the OATP system. The new E3S probe can be used to study the OATP system and as a platform to create radiopharmaceuticals for imaging breast cancer.


Subject(s)
Breast Neoplasms/pathology , Estrone/analogs & derivatives , Hormones/metabolism , Organic Anion Transporters/metabolism , Cell Line, Tumor , Chemistry Techniques, Synthetic , Estrone/chemical synthesis , Estrone/metabolism , Humans , Iodine Radioisotopes , Kinetics , Positron-Emission Tomography , Protein Isoforms/metabolism , Protein Transport , Tomography, Emission-Computed, Single-Photon
2.
Breast Cancer Res Treat ; 145(3): 647-61, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24831777

ABSTRACT

Two-thirds of newly diagnosed hormone-dependent (HR?) breast cancers are detected in post-menopausal patients where estrone-3-sulphate (E3S) is the predominant source for tumour estradiol. Understanding intra-tumoral fate of E3S would facilitate in the identification of novel molecular targets for HR? post-menopausal breast cancer patients. Hence this study investigates the clinical expression of (i) organic anion-transporting polypeptides (OATPs), (ii) multidrug resistance protein (MRP-1), breast cancer resistance proteins (BCRP), and (iii) sulphatase (STS), 17ß-hydroxysteroid dehydrogenase (17ß-HSD-1), involved in E3S uptake, efflux and metabolism, respectively. Fluorescent and brightfield images of stained tumour sections (n = 40) were acquired at 4× and 20× magnification, respectively. Marker densities were measured as the total area of positive signal divided by the surface area of the tumour section analysed and was reported as % area (ImageJ software). Tumour, stroma and non-tumour tissue areas were also quantified (Inform software), and the ratio of optical intensity per histologic area was reported as % area/tumour, % area/stroma and % area/non-tumour. Functional role of OATPs and STS was further investigated in HR? (MCF-7, T47-D, ZR-75) and HR-(MDA-MB-231) cells by transport studies conducted in the presence or absence of specific inhibitors. Amongst all the transporters and enzymes, OATPs and STS have significantly (p < 0.0001) higher expression in HR? tumour sections with highest target signals obtained from the tumour regions of the tissues. Specific OATP-mediated E3S uptake and STS-mediated metabolism were also observed in all HR? breast cancer cells. These observations suggest the potential of OATPs as novel molecular targets for HR? breast cancers.


Subject(s)
Breast Neoplasms/pathology , Estrone/analogs & derivatives , Membrane Transport Proteins/biosynthesis , 17-Hydroxysteroid Dehydrogenases/biosynthesis , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/biosynthesis , Cell Line, Tumor , Estrone/metabolism , Female , Humans , MCF-7 Cells , Multidrug Resistance-Associated Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Organic Anion Transporters/biosynthesis
3.
PLoS One ; 8(5): e64069, 2013.
Article in English | MEDLINE | ID: mdl-23717534

ABSTRACT

The current study investigates the potential of estrone-3-sulphate (E3S) as a ligand for targeting Organic Anion Transporting Polypeptides (OATP), a family of membrane associated uptake transporters, for detection and diagnosis of hormone dependent breast cancers. E3S, an OATP substrate, is a predominant source of tumour estradiol in post-menopausal patients. To assess the potential of E3S as a ligand, distribution of exogenous E3S was determined at the whole body, tumour and cellular levels in murine models of hormone-dependent (MCF-7) and independent (MDA-MB-231) breast cancers. The highest levels of tumour uptake were observed at 6 h post injection (p.i) with significant difference (p = 0.04) between the level in MCF-7 (13.9±3.1%ID/g) and MDA-MB-231 (10.4±1.1%ID/g) (%ID/g: percentage of the total injected dose per gram tissue). The highest tumour-to-blood ratios (MCF-7∶7.4±1.2; MDA-MB-231∶9.1±2.1) were observed at 48 p.i., and highest tumour-to-muscle ratios (MCF-7∶10.7±1.5; MDA-MB-231∶3.8±0.7) were observed at 6 h p.i. Analogous to total tumour uptake, ex vivo tumour cell uptake at 2 h p.i. was 6 fold higher in MCF-7 in comparison to MDA-MB-231 tumour cells. Blocking studies, conducted by pre-administration of 100-fold excess E3S, resulted in significantly lower (MCF-7: p = 0.01; MDA-MB-231: p = 0.02) tumour uptake in both xenograft models, suggesting the involvement of an active carrier-mediated process. The expression of OATP1A2 was detected in tumour sections from both xenografts, with significantly higher expression (p = 0.002) in the MCF-7 xenografts. Overall, the higher tumour uptake and tumour-to-muscle ratio, alongside the higher expression of OATP1A2, in the MCF-7 xenograft model suggests the potential of E3S to serve as a novel ligand for targeting hormone dependent breast cancers.


Subject(s)
Breast Neoplasms/drug therapy , Estrone/analogs & derivatives , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Estradiol/metabolism , Estrone/metabolism , Estrone/pharmacokinetics , Estrone/pharmacology , Female , Humans , Ligands , MCF-7 Cells , Mice , Mice, Nude , Organic Cation Transport Proteins/metabolism
4.
J Pharmacol Exp Ther ; 342(2): 510-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22588260

ABSTRACT

The purpose of this study was to investigate the differential expression and function of organic anion-transporting polypeptides (OATPs) in breast epithelial and breast cancer cells. Estrone-3-sulfate (E3S), a substrate for 7 of 11 OATPs, is a predominant source of tumor estrogen in postmenopausal, hormone-dependent patients with breast cancer. Overexpression of certain OATPs (e.g., OATP1A2) reported in breast tumor tissues compared with surrounding normal tissues could contribute toward two to three times higher tumoral E3S concentration. Little is known about expression and function of other OATP family members among breast epithelial and breast cancer cells. We therefore compared gene and protein expression of seven OATPs (OATP1A2, OATP1B1, OATP1B3, OATP1C1, OATP2B1, OATP3A1, and OATP4A1) in immortalized breast epithelial cells (MCF10A), hormone-dependent breast cancer cells (MCF7), and hormone-independent breast cancer cells (MDA/LCC6-435, MDA-MB-231, and MDA-MB-468) by quantitative polymerase chain reaction and immunoblotting, respectively. Expression of solute carrier superfamily encoding for OATPs (SLCO) 1A2, 1B1, 1B3, 2B1, and 3A1 is exclusive, similar, or significantly higher in cancer cells compared with MCF10A cells. Protein expression of OATPs is found to be either exclusive or higher in cancer cells compared with MCF10A cells. Specificity of OATP-mediated E3S uptake is observed only in cancer cells, with the highest total uptake in MCF7 cells. Transport kinetics of E3S uptake demonstrates transport efficiency that is 10 times greater in the MCF7 cells than in the hormone-independent cells. These data suggest that OATPs could be a novel therapeutic target for hormone-dependent breast cancers, particularly in postmenopausal patients, where the major source of tumor estrogen is E3S.


Subject(s)
Breast Neoplasms/metabolism , Epithelial Cells/metabolism , Estrone/analogs & derivatives , Organic Anion Transporters/metabolism , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Dogs , Estrogens/metabolism , Estrone/genetics , Estrone/metabolism , Female , HEK293 Cells , Humans , Organic Anion Transporters/genetics , Postmenopause/genetics , Postmenopause/metabolism , RNA, Messenger/genetics
5.
Adv Exp Med Biol ; 763: 20-69, 2012.
Article in English | MEDLINE | ID: mdl-23397618

ABSTRACT

Drug transport in the central nervous system can be highly regulated by the expression of numerous influx and efflux transport proteins not only at the blood-brain barrier and blood-cerebrospinal fluid barrier but also in brain parenchymal cellular compartments (i.e., astrocytes, microglia, neurons). In particular, members of the ATP-Binding Cassette membrane-associated transporter superfamily and Solute Carrier family are known to be involved in the traffic of several endobiotics and xenobiotics (including drugs) into and out ofthe brain. These transport proteins have also been implicated in a number of neurological disorders including HIV-encephalitis, Alzheimer's disease, Parkinson's disease and neoplasia. This chapter summarizes recent knowledge on the role of drug transporters in the brain.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Blood-Brain Barrier/metabolism , Carrier Proteins/metabolism , Nervous System Diseases/pathology , Transcytosis , AIDS Dementia Complex/metabolism , AIDS Dementia Complex/pathology , AIDS Dementia Complex/virology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Animals , Astrocytes/metabolism , Blood-Brain Barrier/pathology , Carrier Proteins/genetics , Cell Line , Cell Membrane/metabolism , Cerebrospinal Fluid/metabolism , Choroid Plexus/metabolism , Choroid Plexus/pathology , Diffusion , Endothelial Cells/metabolism , Endothelial Cells/pathology , HIV-1/pathogenicity , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nervous System Diseases/metabolism , Neuroglia/metabolism , Neuroglia/pathology , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Xenobiotics/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...