Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cancer Cell ; 27(6): 809-21, 2015 Jun 08.
Article in English | MEDLINE | ID: mdl-25982816

ABSTRACT

Transforming growth factor-ß (TGF-ß) regulates the expression of genes supporting breast cancer cells in bone, but little is known about prostate cancer bone metastases and TGF-ß. Our study reveals that the TGFBR1 inhibitor SD208 effectively reduces prostate cancer bone metastases. TGF-ß upregulates in prostate cancer cells a set of genes associated with cancer aggressiveness and bone metastases, and the most upregulated gene was PMEPA1. In patients, PMEPA1 expression decreased in metastatic prostate cancer and low Pmepa1 correlated with decreased metastasis-free survival. Only membrane-anchored isoforms of PMEPA1 interacted with R-SMADs and ubiquitin ligases, blocking TGF-ß signaling independently of the proteasome. Interrupting this negative feedback loop by PMEPA1 knockdown increased prometastatic gene expression and bone metastases in a mouse prostate cancer model.


Subject(s)
Bone Neoplasms/secondary , Membrane Proteins/metabolism , Prostatic Neoplasms/pathology , Transforming Growth Factor beta/metabolism , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/prevention & control , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Disease Models, Animal , Gene Knockdown Techniques , Hep G2 Cells , Humans , Male , Membrane Proteins/genetics , Mice , Mice, Nude , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Pteridines/pharmacology , Signal Transduction , Transforming Growth Factor beta/antagonists & inhibitors
2.
ACS Nano ; 5(12): 9480-93, 2011 Dec 27.
Article in English | MEDLINE | ID: mdl-22032622

ABSTRACT

The clinical application of cis-diamminedichloroplatinum(II) (DDP, cisplatin) for cancer therapy is limited by its nonspecific biodistribution and severe side effects. Here, we have developed EGFR-targeted heparin-DDP (EHDDP) nanoparticles for tumor-targeted delivery of DDP. This nanoparticle delivery system possesses the following unique properties: (i) succinic anhydride-modified heparin is biocompatible and biodegradable with no anticoagulant activity; (ii) single-chain variable fragment anti-EGFR antibody (ScFvEGFR) was conjugated to the nanoparticles as an EGFR-targeting ligand. Our results showed that EHDDP nanoparticles can significantly increase the intracellular concentrations of DDP and Pt-DNA adducts in EGFR-expressing non-small cell lung cancer H292 cells via an EGFR-mediated pathway. Compared to the free DDP, significantly prolonged blood circulation time and improved pharmacokinetics and biodistribution of Pt were observed after systemic delivery of the EHDDP nanoparticles. The new EHDDP nanoparticle delivery system significantly enhanced antitumor activity of DDP without weight loss or damage to the kidney and spleen in nude mice bearing H292 cell tumors.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cisplatin/administration & dosage , ErbB Receptors/pharmacokinetics , Heparin/pharmacokinetics , Nanocapsules/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cisplatin/chemistry , Drug Delivery Systems/methods , Humans , Immunoglobulin Variable Region/metabolism , Mice , Mice, Nude , Nanocapsules/chemistry , Treatment Outcome
3.
Curr Top Med Chem ; 10(12): 1211-26, 2010.
Article in English | MEDLINE | ID: mdl-20388107

ABSTRACT

Targeted imaging of cancer is crucial to modern-day cancer management. This review summarizes the current status and future prospects of targeted cancer imaging with MRI, PET, SPECT, CT, and optical imaging techniques. It describes various approaches of cancer imaging and therapy, based on targeting of integrins, somatostatin receptor, epidermal growth factor receptor (EGFR), Her-2/neu receptor, glucose transporter (GLUT), folate receptor, steroid receptor. It also discusses the applications of nanotechnology in imaging and therapy of cancer. Techniques for imaging of cancer in multiple modalities, using a single agent in a single session, have been developed, and this technique is known as 'multimodality imaging'. In order to develop target-specific imaging probes, various targeting ligands, such as small molecules, antibodies, peptides and aptamers have been used. These new imaging agents will help to develop cancer imaging probes that are highly target specific, biocompatible, have high sensitivity, give high signal to noise ratio, and have optimum pharmacokinetic and pharmacodynamic profiles. In another approach, novel agents have been synthesized, suitable for use in imaging as well as in therapy, and they are known as 'theragnostic (or theranostic) agents'. Multidisciplinary approaches and collaborative research efforts from chemists, biologists, biomedical engineers, pharmaceutical scientists, and medical doctors will lead to the discovery of clinically useful imaging and therapeutic agents that can diagnose, prevent, and cure cancer.


Subject(s)
Molecular Probes , Nanomedicine/methods , Neoplasms/diagnosis , Chemistry, Pharmaceutical , Diagnostic Imaging , Humans , Molecular Probes/therapeutic use , Neoplasms/drug therapy
4.
PLoS One ; 4(9): e6896, 2009 Sep 03.
Article in English | MEDLINE | ID: mdl-19727403

ABSTRACT

BACKGROUND: Most patients with advanced breast cancer develop bone metastases, which cause pain, hypercalcemia, fractures, nerve compression and paralysis. Chemotherapy causes further bone loss, and bone-specific treatments are only palliative. Multiple tumor-secreted factors act on the bone microenvironment to drive a feed-forward cycle of tumor growth. Effective treatment requires inhibiting upstream regulators of groups of prometastatic factors. Two central regulators are hypoxia and transforming growth factor (TGF)- beta. We asked whether hypoxia (via HIF-1alpha) and TGF-beta signaling promote bone metastases independently or synergistically, and we tested molecular versus pharmacological inhibition strategies in an animal model. METHODOLOGY/PRINCIPAL FINDINGS: We analyzed interactions between HIF-1alpha and TGF-beta pathways in MDA-MB-231 breast cancer cells. Only vascular endothelial growth factor (VEGF) and the CXC chemokine receptor 4 (CXCR4), of 16 genes tested, were additively increased by both TGF-beta and hypoxia, with effects on the proximal promoters. We inhibited HIF-1alpha and TGF-beta pathways in tumor cells by shRNA and dominant negative receptor approaches. Inhibition of either pathway decreased bone metastasis, with no further effect of double blockade. We tested pharmacologic inhibitors of the pathways, which target both the tumor and the bone microenvironment. Unlike molecular blockade, combined drug treatment decreased bone metastases more than either alone, with effects on bone to decrease osteoclastic bone resorption and increase osteoblast activity, in addition to actions on tumor cells. CONCLUSIONS/SIGNIFICANCE: Hypoxia and TGF-beta signaling in parallel drive tumor bone metastases and regulate a common set of tumor genes. In contrast, small molecule inhibitors, by acting on both tumor cells and the bone microenvironment, additively decrease tumor burden, while improving skeletal quality. Our studies suggest that inhibitors of HIF-1alpha and TGF-beta may improve treatment of bone metastases and increase survival.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia , Transforming Growth Factor beta/metabolism , Animals , Bone Neoplasms/metabolism , Bone and Bones/pathology , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mice , Neoplasm Metastasis , Receptors, CXCR4/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
5.
Clin Cancer Res ; 15(14): 4722-32, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19584158

ABSTRACT

PURPOSE: Cell-surface receptor-targeted magnetic iron oxide nanoparticles provide molecular magnetic resonance imaging contrast agents for improving specificity of the detection of human cancer. EXPERIMENTAL DESIGN: The present study reports the development of a novel targeted iron oxide nanoparticle using a recombinant peptide containing the amino-terminal fragment of urokinase-type plasminogen activator (uPA) conjugated to magnetic iron oxide nanoparticles amino-terminal fragment conjugated-iron oxide (ATF-IO). This nanoparticle targets uPA receptor, which is overexpressed in breast cancer tissues. RESULTS: ATF-IO nanoparticles are able to specifically bind to and be internalized by uPA receptor-expressing tumor cells. Systemic delivery of ATF-IO nanoparticles into mice bearing s.c. and i.p. mammary tumors leads to the accumulation of the particles in tumors, generating a strong magnetic resonance imaging contrast detectable by a clinical magnetic resonance imaging scanner at a field strength of 3 tesla. Target specificity of ATF-IO nanoparticles showed by in vivo magnetic resonance imaging is further confirmed by near-IR fluorescence imaging of the mammary tumors using near-IR dye-labeled amino-terminal fragment peptides conjugated to iron oxide nanoparticles. Furthermore, mice administered ATF-IO nanoparticles exhibit lower uptake of the particles in the liver and spleen compared with those receiving nontargeted iron oxide nanoparticles. CONCLUSIONS: Our results suggest that uPA receptor-targeted ATF-IO nanoparticles have potential as molecularly targeted, dual modality imaging agents for in vivo imaging of breast cancer.


Subject(s)
Magnetic Resonance Imaging/methods , Mammary Neoplasms, Experimental/diagnostic imaging , Nanoparticles/administration & dosage , Receptors, Urokinase Plasminogen Activator/antagonists & inhibitors , Animals , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Ferric Compounds/chemistry , Humans , Image Enhancement/methods , Magnetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Radiography , Receptors, Urokinase Plasminogen Activator/metabolism , Spectroscopy, Near-Infrared
6.
Nanotechnology ; 20(22): 225102, 2009 Jun 03.
Article in English | MEDLINE | ID: mdl-19433879

ABSTRACT

Nanoparticle quantum dots (QDs) provide sharper and more photostable fluorescent signals than organic dyes, allowing quantification of multiple biomarkers simultaneously. In this study, we quantified the expression of epidermal growth factor receptor (EGFR) and E-cadherin (E-cad) in the same cells simultaneously by using secondary antibody-conjugated QDs with two different emission wavelengths (QD605 and QD565) and compared the cellular distribution of EGFR and E-cad between EGFR-tyrosine kinase inhibitor (TKI)-insensitive and -sensitive lung and head and neck cancer cell lines. Relocalization of EGFR and E-cad upon treatment with the EGFR-TKI erlotinib in the presence of EGF was visualized and analyzed quantitatively. Our results showed that QD-immunocytochemistry (ICC)-based technology can not only quantify basal levels of multiple biomarkers but also track the localization of the biomarkers upon biostimulation. With this new technology we found that in EGFR-TKI-insensitive cells, EGFR and E-cad were located mainly in the cytoplasm; while in sensitive cells, they were found mainly on the cell membrane. After induction with EGF, both EGFR and E-cad internalized to the cytoplasm, but the internalization capability in sensitive cells was greater than that in insensitive cells. Quantification also showed that inhibition of EGF-induced EGFR and E-cad internalization by erlotinib in the sensitive cells was stronger than that in the insensitive cells. These studies demonstrate substantial differences between EGFR-TKI-insensitive and -sensitive cancer cells in EGFR and E-cad expression and localization both at the basal level and in response to EGF and erlotinib. QD-based analysis facilitates the understanding of the features of EGFR-TKI-insensitive versus -sensitive cancer cells and may be used in the prediction of patient response to EGFR-targeted therapy.


Subject(s)
Cadherins/metabolism , ErbB Receptors/metabolism , Immunohistochemistry/methods , Neoplasms/metabolism , Quantum Dots , Quinazolines/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Endosomes/metabolism , Erlotinib Hydrochloride , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/metabolism , Humans , Lung Neoplasms/metabolism , Lysosomes/metabolism , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction/drug effects
7.
PLoS One ; 4(4): e5275, 2009.
Article in English | MEDLINE | ID: mdl-19357790

ABSTRACT

During development, growth factors and hormones cooperate to establish the unique sizes, shapes and material properties of individual bones. Among these, TGF-beta has been shown to developmentally regulate bone mass and bone matrix properties. However, the mechanisms that control postnatal skeletal integrity in a dynamic biological and mechanical environment are distinct from those that regulate bone development. In addition, despite advances in understanding the roles of TGF-beta signaling in osteoblasts and osteoclasts, the net effects of altered postnatal TGF-beta signaling on bone remain unclear. To examine the role of TGF-beta in the maintenance of the postnatal skeleton, we evaluated the effects of pharmacological inhibition of the TGF-beta type I receptor (TbetaRI) kinase on bone mass, architecture and material properties. Inhibition of TbetaRI function increased bone mass and multiple aspects of bone quality, including trabecular bone architecture and macro-mechanical behavior of vertebral bone. TbetaRI inhibitors achieved these effects by increasing osteoblast differentiation and bone formation, while reducing osteoclast differentiation and bone resorption. Furthermore, they induced the expression of Runx2 and EphB4, which promote osteoblast differentiation, and ephrinB2, which antagonizes osteoclast differentiation. Through these anabolic and anti-catabolic effects, TbetaRI inhibitors coordinate changes in multiple bone parameters, including bone mass, architecture, matrix mineral concentration and material properties, that collectively increase bone fracture resistance. Therefore, TbetaRI inhibitors may be effective in treating conditions of skeletal fragility.


Subject(s)
Bone and Bones/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism , Animals , Bone Density/drug effects , Bone Development/drug effects , Bone Matrix/metabolism , Bone Resorption/metabolism , Bone and Bones/anatomy & histology , Bone and Bones/cytology , Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Receptor, EphB4/metabolism , Receptor, Transforming Growth Factor-beta Type I
8.
Int J Nanomedicine ; 3(3): 311-21, 2008.
Article in English | MEDLINE | ID: mdl-18990940

ABSTRACT

Magnetic iron oxide (IO) nanoparticles with a long blood retention time, biodegradability and low toxicity have emerged as one of the primary nanomaterials for biomedical applications in vitro and in vivo. IO nanoparticles have a large surface area and can be engineered to provide a large number of functional groups for cross-linking to tumor-targeting ligands such as monoclonal antibodies, peptides, or small molecules for diagnostic imaging or delivery of therapeutic agents. IO nanoparticles possess unique paramagnetic properties, which generate significant susceptibility effects resulting in strong T2 and T*2 contrast, as well as T1 effects at very low concentrations for magnetic resonance imaging (MRI), which is widely used for clinical oncology imaging. We review recent advances in the development of targeted IO nanoparticles for tumor imaging and therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Ferric Compounds/administration & dosage , Magnetic Resonance Imaging/methods , Nanoparticles/administration & dosage , Neoplasms/diagnosis , Neoplasms/drug therapy , Contrast Media , Drug Delivery Systems/methods , Electromagnetic Fields , Humans , Image Enhancement/methods
9.
Nat Biotechnol ; 26(1): 83-90, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18157119

ABSTRACT

We describe biocompatible and nontoxic nanoparticles for in vivo tumor targeting and detection based on pegylated gold nanoparticles and surface-enhanced Raman scattering (SERS). Colloidal gold has been safely used to treat rheumatoid arthritis for 50 years, and has recently been found to amplify the efficiency of Raman scattering by 14-15 orders of magnitude. Here we show that large optical enhancements can be achieved under in vivo conditions for tumor detection in live animals. An important finding is that small-molecule Raman reporters such as organic dyes were not displaced but were stabilized by thiol-modified polyethylene glycols. These pegylated SERS nanoparticles were considerably brighter than semiconductor quantum dots with light emission in the near-infrared window. When conjugated to tumor-targeting ligands such as single-chain variable fragment (ScFv) antibodies, the conjugated nanoparticles were able to target tumor biomarkers such as epidermal growth factor receptors on human cancer cells and in xenograft tumor models.


Subject(s)
Carcinoma, Squamous Cell/diagnosis , Drug Delivery Systems/methods , Nanoparticles , Spectrum Analysis, Raman/methods , Animals , Cell Line, Tumor , Humans , Mice , Particle Size
10.
Mol Pharmacol ; 71(1): 101-11, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17035597

ABSTRACT

The identification of differentially regulated apoptotic signals in normal and tumor cells allows the development of cancer cell-selective therapies. Increasing evidence shows that the inhibitor of apoptosis (IAP) proteins survivin and XIAP are highly expressed in tumor cells but are absent or have very low levels of expression in normal adult tissues. We found that inhibiting AKT activity with 10 to 100 nM deguelin, a small molecule derived from natural products, markedly reduced the levels of both survivin and XIAP, inducing apoptosis in human breast cancer cells but not in normal cells. It is noteworthy that we detected an elevated level of cleaved poly(ADP-ribose) polymerase, a signature of caspase activation, without a significant increase in caspase activity in deguelin-treated cancer cells. Our results suggest that severe down-regulation of the IAPs by deguelin releases their inhibitory activity over pre-existing active caspases present in cancer cells, inducing apoptosis without the need for further caspase activation. Because normal cells have very low levels of p-AKT, XIAP, survivin, and pre-existing caspase activity, deguelin had little effect on those cells. In addition, we found that combining deguelin with chemotherapy drugs enhanced drug-induced apoptosis selectively in human tumor cells, which suggests that deguelin has great potential for chemosensitization and could represent a new therapeutic agent for treatment of breast cancer.


Subject(s)
Inhibitor of Apoptosis Proteins/genetics , Rotenone/analogs & derivatives , X-Linked Inhibitor of Apoptosis Protein/genetics , Apoptosis , Breast Neoplasms/pathology , Cell Division , Cell Line, Tumor , Colony-Forming Units Assay , DNA Primers , Down-Regulation/drug effects , Female , Genes, Reporter , Humans , Inhibitor of Apoptosis Proteins/drug effects , Inhibitor of Apoptosis Proteins/metabolism , Microtubule-Associated Proteins/drug effects , Microtubule-Associated Proteins/genetics , Neoplasm Proteins/drug effects , Neoplasm Proteins/genetics , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rotenone/pharmacology , Survivin , X-Linked Inhibitor of Apoptosis Protein/drug effects , X-Linked Inhibitor of Apoptosis Protein/metabolism
11.
J Biol Chem ; 281(36): 25903-14, 2006 Sep 08.
Article in English | MEDLINE | ID: mdl-16847054

ABSTRACT

Although increasing evidence supports a link between epidermal growth factor receptor (EGFR) signaling and resistance to apoptosis, the mechanism by which the EGFR signaling pathway inhibits apoptosis is not well understood. In this study, we found that epidermal growth factor (EGF) stimulation increased the level of expression of the inhibitor of apoptosis protein survivin in breast cancer cells but not in normal mammary epithelial cells. We further demonstrated that activation of survivin gene expression is mediated by oxygen-independent hypoxia-inducible factor (HIF)-1alpha up-regulation in EGF-treated cancer cells. EGFR signaling activated the phosphoinositide 3-kinase/AKT pathway, subsequently increasing the level of HIF-1alpha under normoxic conditions. HIF-1alpha then activated survivin gene transcription through direct binding to the survivin promoter. Furthermore, we found that overexpression of HIF-1alpha small interfering RNA blocks EGF-induced survivin gene up-regulation and increases apoptosis induced by the chemotherapy drug docetaxel. However, transfection of a plasmid expressing HIF-1alpha gene activates survivin gene expression and reduces the apoptotic response. Our results demonstrate a novel pathway for EGFR signaling-mediated apoptosis resistance in human cancer cells. Although the role of HIF-1alpha in regulating cell survival under hypoxic conditions has been studied extensively, our results show that normoxic breast cancer cells utilize cross-talk between EGFR signals and HIF-1alpha to up-regulate the anti-apoptotic survivin gene, providing a strong rationale for the targeting of HIF-1alpha as a therapeutic approach for both hypoxic and normoxic tumor cells. Understanding key molecular events in EGFR signaling-induced apoptosis resistance should provide new information for the development of novel therapeutic agents targeting EGFR, HIF-1alpha, and/or survivin.


Subject(s)
Apoptosis/physiology , ErbB Receptors/metabolism , Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Signal Transduction/physiology , Antineoplastic Agents, Phytogenic/metabolism , Breast Neoplasms , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation , Docetaxel , Enzyme Inhibitors/metabolism , Epidermal Growth Factor/metabolism , ErbB Receptors/genetics , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/genetics , Neoplasm Proteins/genetics , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/metabolism , Survivin , Taxoids/metabolism
12.
Biochemistry ; 45(24): 7605-16, 2006 Jun 20.
Article in English | MEDLINE | ID: mdl-16768456

ABSTRACT

Each nucleotide-binding domain (NBD) of mammalian P-glycoproteins (Pgps) and human ATP-binding cassette (ABC) B subfamily members contains a tyrosine residue approximately 25 residues upstream of the Walker A domain. To assess the role of the conserved Y401 and Y1044 residues of human Pgp, we substituted these residues with F, W, C, or A either singly or together. The mutant proteins were expressed in a Vaccinia virus-based transient expression system as well as in baculovirus-infected HighFive insect cells. The Y401F, Y401W, Y1044F, Y1044W, or Y401F/Y1004F mutants transported fluorescent substrates similar to the wild-type protein. On the other hand, Y401L and Y401C exhibited partial (30-50%) function, and transport was completely abolished in Y401A, Y1044A, and Y401A/Y1044A mutant Pgps. Similarly, in Y401A, Y1044A, and Y401A/Y1044A mutants, TNP-ATP binding, vanadate-induced trapping of nucleotide, and ATP hydrolysis were completely abolished. Thus, an aromatic residue upstream of the Walker A motif in ABC transporters is critical for binding of ATP. Additionally, the crystal structures of several NBDs in the nucleotide-bound form, data mining, and alignment of 18,514 ABC domains with the consensus conserved sequence in a database of all nonredundant proteins indicate that an aromatic residue is highly conserved in approximately 85% of ABC proteins. Although the role of this aromatic residue has previously been studied in a few ABC proteins, we provide evidence for a near-universal structural and functional role for this residue and recognize its presence as a conserved subdomain approximately 25 amino acids upstream of the Walker A motif that is critical for ATP binding. We named this subdomain the "A-loop" (aromatic residue interacting with the adenine ring of ATP).


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , ATP-Binding Cassette Transporters/chemistry , Adenosine Triphosphate/metabolism , Tyrosine/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP-Binding Cassette Transporters/genetics , Adenosine Triphosphatases/analysis , Adenosine Triphosphate/chemistry , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Animals , Baculoviridae/genetics , Base Sequence , Conserved Sequence , Dimerization , HeLa Cells , Humans , Hydrolysis , Insecta/cytology , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Nucleic Acid , Trypsin/pharmacology , Vaccinia virus/metabolism
13.
Cancer Res ; 65(5): 1909-17, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15753390

ABSTRACT

Development of novel approaches for quantitative analysis of gene expression in intact tumor cells should provide new means for cancer detection and for studying the response of cancer cells to biological and therapeutic reagents. We developed procedures for detecting the levels of expression of multiple genes in fixed as well as viable cells using molecular beacon imaging technology. We found that simultaneous delivery of molecular beacons targeting survivin and cyclin D1 mRNAs produced strong fluorescence in breast cancer but not in normal breast cells. Importantly, fluorescence intensity correlated well with the level of gene expression in the cells detected by real-time reverse transcription-PCR or Western blot analysis. We further show that molecular beacons can detect changes of survivin gene expression in viable cancer cells following epidermal growth factor stimulation, docetaxel treatment, and overexpression of p53 gene. Thus, molecular beacon imaging is a simple and specific method for detecting gene expression in cancer cells. It has great potential for cancer detection and drug development.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/diagnosis , Microtubule-Associated Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/metabolism , Base Sequence , Biomarkers, Tumor/genetics , Blotting, Western , Breast/metabolism , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Female , Fluorescent Dyes/metabolism , Humans , Image Interpretation, Computer-Assisted , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/genetics , Molecular Probes , Molecular Sequence Data , Neoplasm Proteins , RNA, Messenger/metabolism , Sensitivity and Specificity , Sequence Homology, Nucleic Acid , Survivin , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
14.
Mol Pharmacol ; 65(3): 675-84, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14978246

ABSTRACT

The overexpression of multidrug resistance protein 1 (MDR1) and multidrug resistance protein 1 (MRP1) gene products is a major cause of multidrug resistance in cancer cells. A recent study suggested that disulfiram, a drug used to treat alcoholism, might act as a modulator of P-glycoprotein. In this study, we investigated the molecular and chemical basis of disulfiram as a multidrug resistance modulator. We demonstrate that in intact cells, disulfiram reverses either MDR1- or MRP1-mediated efflux of fluorescent drug substrates. Disulfiram inhibits ATP hydrolysis and the binding of [alpha-32P]8-azidoATP to P-glycoprotein and MRP1, with inhibition curves comparable with those of N-ethylmaleimide, a cysteine-modifying agent. However, if the ATP sites are protected with excess ATP, disulfiram stimulates ATP hydrolysis by both transporters in a concentration-dependent manner. Thus, in addition to modifying cysteines at the ATP sites, disulfiram may interact with the drug-substrate binding site. We demonstrate that disulfiram, but not N-ethylmaleimide, inhibits in a concentration-dependent manner the photoaffinity labeling of the multidrug transporter with 125I-iodoarylazidoprazosin and [3H]azidopine. This suggests that the interaction of disulfiram with the drug-binding site is independent of its role as a cysteine-modifying agent. Finally, we have exploited MRP4 (ABCC4) to demonstrate that disulfiram can inhibit ATP binding by forming disulfide bonds between cysteines located in the vicinity of, although not in, the active site. Taken together, our results suggest that disulfiram has unique molecular interactions with both the ATP and/or drug-substrate binding sites of multiple ATP binding cassette transporters, which are associated with drug resistance, and it is potentially an attractive agent to combat multidrug resistance.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adenosine Triphosphate/analogs & derivatives , Disulfiram/pharmacology , Drug Resistance, Multiple/physiology , Multidrug Resistance-Associated Proteins/metabolism , Prazosin/analogs & derivatives , 3T3 Cells , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Azides/pharmacology , Binding Sites/drug effects , Cells, Cultured , Dihydropyridines/pharmacology , Drug Interactions , Enzyme Inhibitors/pharmacology , Humans , Hydrolysis , Mice , Phosphorus Radioisotopes , Prazosin/pharmacology
15.
Biochemistry ; 41(47): 13989-4000, 2002 Nov 26.
Article in English | MEDLINE | ID: mdl-12437356

ABSTRACT

The human MDR1 (ABCB1) gene product, P-glycoprotein (Pgp), functions as an ATP-dependent efflux pump for a variety of chemotherapeutic drugs. In this study, we assessed the role of conserved glutamate residues in the Walker B domain of the two ATP sites (E556 and E1201, respectively) during the catalytic cycle of human Pgp. The mutant Pgps (E556Q, E556A, E1201Q, E1201A, E556/1201Q, and E556/1201A) were characterized using a vaccinia virus based expression system. Although steady-state ATP hydrolysis and drug transport activities were abrogated in both E556Q and E1201Q mutant Pgps, [alpha-(32)P]-8-azidoADP was trapped in the presence of vanadate (Vi), and the release of trapped [alpha-(32)P]-8-azidoADP occurred to a similar extent as in wild-type Pgp. This indicates that these mutations do not affect either the first hydrolysis event or the ADP release step. Similar results were also obtained when Glu residues were replaced with Ala (E556A and E1201A). Following the first hydrolysis event and release of [alpha-(32)P]-8-azidoADP, both E556Q and E1201Q mutant Pgps failed to undergo another cycle of Vi-induced [alpha-(32)P]-8-azidoADP trapping. Interestingly, the double mutants E556/1201Q and E556/1201A trapped [alpha-(32)P]-8-azidoADP even in the absence of Vi, and the occluded nucleotide was not released after incubation at 37 degrees C for an extended period. In addition, the properties of transition state conformation of the double mutants generated in the absence of Vi were found to be similar to that of the wild-type protein trapped in the presence of Vi (Pgp x [alpha-(32)P]-8-azidoADP xVi). Thus, in contrast to the single mutants, the double mutants appear to be defective in the ADP release step. In aggregate, these data suggest that E556 and E1201 residues in the Walker B domains may not be critical as catalytic carboxylates for the cleavage of the bond between the gamma-P and the beta-P of ATP during hydrolysis but are essential for the second ATP hydrolysis step and completion of the catalytic cycle.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adenosine Triphosphate/metabolism , Glutamic Acid , ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , Catalysis , Conserved Sequence , DNA Primers , Humans , Hydrolysis , Kinetics , Mutagenesis, Site-Directed , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Vanadates/pharmacology
16.
Biochemistry ; 41(31): 10123-32, 2002 Aug 06.
Article in English | MEDLINE | ID: mdl-12146977

ABSTRACT

To enable cell surface localization of the human multidrug resistance protein (MRP1, ABCC1) and to assess the role of the extracellular domains of this transporter, the FLAG epitope tag was introduced into different extracellular loops of the three membrane-spanning domains (MSDs) of the transporter. We constructed and expressed various partially and fully glycosylation-deficient, FLAG-tagged MRP1 proteins in a Vaccinia virus-based transient expression system, and the cell surface expression level of MRP1 on intact cells was followed by flow cytometry, using the FLAG tag specific monoclonal antibody M2. We also expressed the wild-type MRP1 protein and some of the FLAG-tagged mutants in stably transfected HEK293 cells, and followed the cell surface expression and the transport function of MRP1 both by monitoring the efflux of fluorescent substrate and by their ability to confer resistance to HEK293 transfectants to anticancer agents such as daunorubicin and etoposide. When we inserted the FLAG epitope in extracellular loops of the MSD1 or MSD3, the tag was accessible upon removal of N-glycosylation sites (N --> Q at positions 17, 23, and 1006, respectively), whereas the FLAG epitope placed in the MSD2 was not accessible even after removal of all three N-glycosylation sites, indicating that MSD2 region is deeply buried in the plasma membrane. However, all FLAG tagged MRP1 mutants were expressed at the cell surface to the same extent as the wild-type protein and also exhibited normal transport function. Our results demonstrate that the accessibility of the external FLAG epitope is strongly dependent on the position of the tag and the glycosylation state of the different FLAG-tagged MRP1s, and the conformation of extracellular loops in MSD1 and MDS3 does not appear to contribute to the functional status of MRP1.


Subject(s)
Multidrug Resistance-Associated Proteins/metabolism , Base Sequence , DNA Primers , Electrophoresis, Polyacrylamide Gel , Glycosylation , HeLa Cells , Humans , Multidrug Resistance-Associated Proteins/chemistry , Mutagenesis, Site-Directed , Plasmids , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...