Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Endocr Relat Cancer ; 24(9): 459-474, 2017 09.
Article in English | MEDLINE | ID: mdl-28676500

ABSTRACT

Pituitary tumor-transforming gene 1-binding factor (PTTG1IP; PBF) is a multifunctional glycoprotein, which is overexpressed in a wide range of tumours, and significantly associated with poorer oncological outcomes, such as early tumour recurrence, distant metastasis, extramural vascular invasion and decreased disease-specific survival. PBF transforms NIH 3T3 fibroblasts and induces tumours in nude mice, while mice harbouring transgenic thyroidal PBF expression show hyperplasia and macrofollicular lesions. Our assumption that PBF becomes an oncogene purely through increased expression has been challenged by the recent report of mutations in PBF within the Catalogue of Somatic Mutations in Cancer (COSMIC) database. We therefore sought to determine whether the first 10 PBF missense substitutions in human cancer might be oncogenic. Anisomycin half-life studies revealed that most mutations were associated with reduced protein stability compared to wild-type (WT) PBF. Proliferation assays narrowed our interest to two mutational events which significantly altered cell turnover: C51R and R140W. C51R was mainly confined to the endoplasmic reticulum while R140W was apparent in the Golgi apparatus. Both C51R and R140W lost the capacity to induce cellular migration and significantly reduced cell invasion. Colony formation and soft agar assays demonstrated that, in contrast to WT PBF, both mutants were unable to elicit significant colony formation or anchorage-independent growth. However, C51R and R140W retained the ability to repress radioiodide uptake, a functional hallmark of PBF. Our data reveal new insight into PBF function and confirm that, rather than being oncogenic, mutations in PBF are likely to be passenger effects, with overexpression of PBF the more important aetiological event in human cancer.


Subject(s)
Membrane Proteins/genetics , Animals , Cell Proliferation , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/metabolism , Mice , Mutation , Proto-Oncogene Mas , Transfection
2.
Oncogene ; 36(37): 5296-5308, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28504713

ABSTRACT

The proto-oncogene PTTG and its binding partner PBF have been widely studied in multiple cancer types, particularly thyroid and colorectal, but their combined role in tumourigenesis is uncharacterised. Here, we show for the first time that together PTTG and PBF significantly modulate DNA damage response (DDR) genes, including p53 target genes, required to maintain genomic integrity in thyroid cells. Critically, DDR genes were extensively repressed in primary thyrocytes from a bitransgenic murine model (Bi-Tg) of thyroid-specific PBF and PTTG overexpression. Irradiation exposure to amplify p53 levels further induced significant repression of DDR genes in Bi-Tg thyrocytes (P=2.4 × 10-4) compared with either PBF- (P=1.5 × 10-3) or PTTG-expressing thyrocytes (P=NS). Consistent with this, genetic instability was greatest in Bi-Tg thyrocytes with a mean genetic instability (GI) index of 35.8±2.6%, as well as significant induction of gross chromosomal aberrations in thyroidal TPC-1 cells following overexpression of PBF and PTTG. We extended our findings to human thyroid cancer using TCGA data sets (n=322) and found striking correlations with PBF and PTTG expression in well-characterised DDR gene panel RNA-seq data. In addition, genetic associations and transient transfection identified PBF as a downstream target of the receptor tyrosine kinase-BRAF signalling pathway, emphasising a role for PBF as a novel component in a pathway well described to drive neoplastic growth. We also showed that overall survival (P=1.91 × 10-5) and disease-free survival (P=4.9 × 10-5) was poorer for TCGA patients with elevated tumoural PBF/PTTG expression and mutationally activated BRAF. Together our findings indicate that PBF and PTTG have a critical role in promoting thyroid cancer that is predictive of poorer patient outcome.


Subject(s)
DNA Damage , Membrane Proteins/metabolism , Securin/metabolism , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Animals , Disease Models, Animal , Female , Humans , Intracellular Signaling Peptides and Proteins , Male , Membrane Proteins/genetics , Mice , Mice, Transgenic , Prognosis , Proto-Oncogene Mas , Securin/genetics , Survival Rate , Thyroid Neoplasms/pathology , Transfection , Treatment Outcome
3.
J Clin Endocrinol Metab ; 98(7): 2876-86, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23678037

ABSTRACT

CONTEXT: The clinical effectiveness of ablative radioiodine treatment of thyroid tumors is limited by the availability of the sodium iodide symporter (NIS) at the plasma membrane (PM) for uptake of ¹³¹I. A significant proportion of well-differentiated thyroid tumors are unable to concentrate sufficient radioiodine for effective therapy, and in other tumor models such as breast tumors, where radioiodine uptake would be an attractive therapeutic option, uptake is insufficient. OBJECTIVE: Pituitary tumor-transforming gene-binding factor (PBF; PTTG1IP) is overexpressed in multiple cancers and significantly decreases NIS expression at the PM. The goal of this study was to identify a method by which PBF repression of NIS may be overcome in human tumors. RESULTS: Here, we identify PBF as a tyrosine phosphoprotein that specifically binds the proto-oncogene tyrosine protein kinase Src in mass spectrometry, glutathione S-transferase pulldown and coimmunoprecipitation assays. Src induction leads to phosphorylation at PBF residue Y174. Abrogation of this residue results in PM retention and a markedly reduced ability to bind NIS. The Src inhibitor PP1 inhibits PBF phosphorylation in multiple cell lines in vitro, including human primary thyroid cells. Of direct clinical importance to the treatment of thyroid cancer, PP1 stimulates iodide uptake by transfected NIS in TPC1 thyroid carcinoma cells and entirely overcomes PBF repression of iodide uptake in human primary thyroid cells. CONCLUSIONS: We propose that targeting PBF phosphorylation at residue Y174 via tyrosine kinase inhibitors may be a novel therapeutic strategy to enhance the efficacy of ablative radioiodine treatment in thyroid and other endocrine and endocrine-related tumors.


Subject(s)
Cell Membrane/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Symporters/metabolism , Thyroid Gland/metabolism , Thyroid Neoplasms/metabolism , Amino Acid Substitution , Animals , Biological Transport/drug effects , COS Cells , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/pathology , Cells, Cultured , Chlorocebus aethiops , Humans , Intracellular Signaling Peptides and Proteins , Iodine Radioisotopes/metabolism , Membrane Proteins/genetics , Mutant Proteins/metabolism , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Mas , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Proto-Oncogene Proteins pp60(c-src)/metabolism , Radiopharmaceuticals/metabolism , Recombinant Proteins/agonists , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Symporters/agonists , Symporters/genetics , Thyroid Gland/cytology , Thyroid Gland/drug effects , Thyroid Gland/pathology , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/pathology , Thyroid Neoplasms/radiotherapy
4.
Endocrinology ; 153(7): 3526-36, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22535767

ABSTRACT

Within the basolateral membrane of thyroid follicular epithelial cells, two transporter proteins are central to thyroid hormone (TH) biosynthesis and secretion. The sodium iodide symporter (NIS) delivers iodide from the bloodstream into the thyroid, and after TH biosynthesis, monocarboxylate transporter 8 (MCT8) mediates TH secretion from the thyroid gland. Pituitary tumor-transforming gene-binding factor (PBF; PTTG1IP) is a protooncogene that is up-regulated in thyroid cancer and that binds NIS and modulates its subcellular localization and function. We now show that PBF binds MCT8 in vitro, eliciting a marked shift in MCT8 subcellular localization and resulting in a significant reduction in the amount of MCT8 at the plasma membrane as determined by cell surface biotinylation assays. Colocalization and interaction between PBF and Mct8 was also observed in vivo in a mouse model of thyroid-specific PBF overexpression driven by a bovine thyroglobulin (Tg) promoter (PBF-Tg). Thyroidal Mct8 mRNA and protein expression levels were similar to wild-type mice. Critically, however, PBF-Tg mice demonstrated significantly enhanced thyroidal TH accumulation and reduced TH secretion upon TSH stimulation. Importantly, Mct8-knockout mice share this phenotype. These data show that PBF binds and alters the subcellular localization of MCT8 in vitro, with PBF overexpression leading to an accumulation of TH within the thyroid in vivo. Overall, these studies identify PBF as the first protein to interact with the critical TH transporter MCT8 and modulate its function in vivo. Furthermore, alongside NIS repression, PBF may thus represent a new regulator of TH biosynthesis and secretion.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation , Thyroid Hormones/metabolism , Animals , Biotinylation , COS Cells , Chlorocebus aethiops , DNA, Complementary/metabolism , Glutathione Transferase/metabolism , Intracellular Signaling Peptides and Proteins , Membrane Transport Proteins/metabolism , Mice , Models, Biological , Monocarboxylic Acid Transporters , Phenotype , Protein Processing, Post-Translational , Symporters , Tetraspanin 30/biosynthesis , Transcription, Genetic
5.
Mol Cell Endocrinol ; 278(1-2): 1-6, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17928133

ABSTRACT

It is now 10 years since PTTG was first cloned and isolated. Perhaps the major story of the intervening decade of work performed by numerous groups around the world is the sheer multifunctionality ascribed to this gene. PTTG has been implicated in mechanisms of gene transactivation, cell transformation, angiogenesis, metabolism, apoptosis, DNA repair, genetic instability and mitotic control, both in endocrine and non-endocrine settings. In the current review, we cast a critical eye over a decade of PTTG research within the field of endocrine neoplasia.


Subject(s)
Cell Transformation, Neoplastic/genetics , Neoplasm Proteins/physiology , Pituitary Neoplasms/genetics , Animals , Humans , Mice , Neoplasm Proteins/genetics , Rats , Securin
6.
J Gene Med ; 6(4): 429-38, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15079817

ABSTRACT

BACKGROUND: Efficient neuronal gene therapy is a goal for the long-term repair and regeneration of the injured central nervous system (CNS). We investigated whether targeting cDNA to neurons with cholera toxin b chain conjugated non-viral polyplexes led to increased efficiency of non-viral gene transfer in the CNS. Here, we illustrate the potential for this strategy by demonstrating enhanced transfection of a differentiated neuronal cell type, PC12. METHODS: In vitro transfection efficiency of a cholera toxin b chain-poly(D-lysine) molecular conjugate (CTb-K(100)) was compared by fluorescence-activated cell sorting (FACS) analysis of green fluorescent protein (GFP) expression and luminometric measurement of beta-galactosidase (beta-gal) expression, to untargeted poly(D-lysine) (K(100)) in undifferentiated and NGF-differentiated PC12 cells. RESULTS: Transfection of undifferentiated PC12 cells with CTb-K(100) polyplexes resulted in a 36-fold increase in levels of pCMV-DNA(LacZ) expression and a 20-fold increase in the frequency of transduction with pCMV-DNA(GFP), compared with untargeted K(100) polyplexes. Treatment of PC12 cells with 50 ng/ml/day of NGF for 14 days led to differentiation to a neuronal phenotype. Transfection of NGF-differentiated cells with CTb-K(100) polyplexes resulted in a 133-fold increase in levels of pCMV-DNA(LacZ) expression and a 11-fold increase in the percentage of cells transduced with pCMV-DNA(GFP), compared with untargeted K(100) polyplexes. Transfection was dependent on CTb, with CTb-K(100)-mediated transfections competitively inhibited with free CTb in both PC12 phenotypes. CONCLUSIONS: Non-viral systems for gene transfer in damaged CNS show superior toxicological profiles to most viruses but are limited by inefficient and non-selective gene expression in target tissue. Cholera toxin is known to interact preferentially with neuronal cells of the central and peripheral nervous systems, mediating binding through the b subunit, CTb, and the pentasaccharide moiety of the gangliosaccharide, GM1, which is present at high levels on the neuronal cell surface. Here, we show that a molecular conjugate of the CTb subunit, covalently linked to poly(D-lysine), is able to successfully target and significantly enhance transfection of a neuronal cell type, NGF-differentiated rat PC12 pheochromocytoma cells. This observation encourages the further development of non-viral strategies for the delivery of therapeutic genes to neurons.


Subject(s)
Cholera Toxin/genetics , Gene Transfer Techniques , Neurons/physiology , Polylysine/genetics , Animals , Binding, Competitive , Cell Differentiation/drug effects , Cell Differentiation/genetics , Chemistry, Physical/methods , Cholera Toxin/metabolism , DNA, Complementary , G(M1) Ganglioside/metabolism , Gene Expression , Nerve Growth Factor/pharmacology , PC12 Cells , Rats , Transfection/methods
7.
Nucleic Acids Res ; 29(18): 3882-91, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11557821

ABSTRACT

Synthetic vectors were evaluated for their ability to mediate efficient mRNA transfection. Initial results indicated that lipoplexes, but not polyplexes based on polyethylenimine (PEI, 25 and 22 kDa), poly(L-lysine) (PLL, 54 kDa) or dendrimers, mediated efficient translation of mRNA in B16-F10 cells. Significant mRNA transfection was achieved by lipoplex delivery in quiescent (passage 0) human umbilical vein endothelial cells (HUVEC), and by passage 4, 10.7% of HUVEC were transfected compared to 0.84% with DNA. Lack of expression with PEI 25 kDa/mRNA or PLL 54 kDa/mRNA in a cell-free translation assay and following cytoplasmic injection into Rat1 cells indicated that these polyplexes were too stable to release mRNA. In contrast, polyplexes formed using smaller PEI 2 kDa and PLL 3.4 kDa gave 5-fold greater expression in B16-F10 cells compared to DOTAP, but were dependent on chloroquine for transfection activity. Endosomolytic activity was incorporated by conjugating PEI 2 kDa to melittin and resulting PEI 2 kDa-melittin/mRNA polyplexes mediated high transfection levels in HeLa cells (31.1 +/- 4.1%) and HUVEC (58.5 +/- 2.9%) in the absence of chloroquine, that was potentiated to 52.2 +/- 2.7 and 71.6 +/- 1.7%, respectively, in the presence of chloroquine. These results demonstrate that mRNA polyplexes based on peptide-modified low molecular weight polycations can possess versatile properties including endosomolysis that should enable efficient non-viral mRNA transfection of quiescent and post-mitotic cells.


Subject(s)
Oligopeptides/physiology , RNA/metabolism , Transfection/methods , Amino Acid Sequence , Animals , Cell Line , Cell-Free System/metabolism , Gene Expression , Green Fluorescent Proteins , HeLa Cells , Humans , Luciferases/genetics , Luciferases/metabolism , Luminescent Proteins/genetics , Melitten/chemistry , Melitten/genetics , Microinjections , Mitosis , Molecular Sequence Data , Oligopeptides/genetics , Protein Biosynthesis , RNA/genetics , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Reticulocytes/chemistry , Reticulocytes/metabolism , Time Factors , Transcription, Genetic , Tumor Cells, Cultured
8.
Curr Opin Mol Ther ; 3(2): 116-24, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11338923

ABSTRACT

The introduction of RNA into mammalian cells is a relatively straightforward procedure with many therapeutic applications. An advantage of using mRNA is that protein expression can be achieved in post-mitotic or quiescent cells where there is usually little or no gene expression with non-viral DNA delivery systems. Furthermore, the cleavage of mRNA by catalytic RNA molecules, or ribozymes, is a useful strategy to downregulate aberrant gene expression. The purpose of this review is to provide an update of current applications that use RNA molecules such as mRNA and ribozymes as a basis for gene therapy strategies targeting the initiation and progression of cancer. In particular, we focus on recent developments that improve the delivery and stability of RNA molecules to achieve therapeutic efficacy.


Subject(s)
Genetic Therapy , Neoplasms/therapy , RNA, Catalytic/therapeutic use , RNA, Messenger/genetics , Animals , Cancer Vaccines , Genetic Vectors , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Nucleic Acid Conformation , RNA Stability , RNA, Catalytic/administration & dosage , RNA, Catalytic/metabolism , RNA, Messenger/metabolism
9.
Blood ; 97(8): 2221-9, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11290582

ABSTRACT

Effective gene therapy for diseases of the circulation requires vectors capable of systemic delivery. The molecular weight of poly(L-lysine) (pLL) has a significant effect on the circulation of pLL/DNA complexes in mice, with pLL(211)/DNA complexes displaying up to 20 times greater levels in the blood after 30 minutes compared with pLL(20)/DNA. It is shown that pLL(20)/DNA complexes fix mouse complement C3 in vitro, independent of immunoglobulin binding; are less soluble in the blood in vivo; bind erythrocytes; are rapidly removed by the liver, where they associate predominantly with Kupffer cells; and result in a rapid increase in hepatic leukocytes expressing high levels of complement receptor 3 (CR3). The circulation properties of these complexes are also dependent on the type of DNA used, with circular plasmid DNA complexes exhibiting increased circulation compared with linear DNA. PLL(211)/DNA complexes bind erythrocytes and associate with Kupffer cells but, in contrast, do not fix mouse complement in vitro and are unaffected by the type of DNA used. In rats, both types of complexes produce hematuria and are rapidly removed from the circulation. Correlation of in vivo and in vitro results suggests that the solubility of complexes in physiological saline and species-matched complement fixation and erythrocyte lysis may correlate with systemic circulation. Analysis using human blood in vitro shows no hemolysis, but both types of complexes fix complement and bind IgG, suggesting that pLL/DNA complexes may be rapidly cleared from the human circulation.


Subject(s)
DNA, Circular/pharmacokinetics , DNA, Recombinant/pharmacokinetics , Genetic Therapy , Genetic Vectors/pharmacokinetics , Polylysine/pharmacokinetics , Animals , Blood Proteins/metabolism , Complement Activation , Complement C3/metabolism , DNA, Circular/blood , DNA, Recombinant/blood , Female , Genetic Vectors/blood , Genetic Vectors/toxicity , Hematuria/chemically induced , Humans , Immunomagnetic Separation , Injections, Intravenous , Kupffer Cells/metabolism , Leukocytes/metabolism , Liver/metabolism , Mice , Mice, Inbred BALB C , Molecular Weight , Polylysine/blood , Polylysine/chemistry , Polylysine/toxicity , Rats , Rats, Wistar , Receptors, Complement/biosynthesis , Solubility , Species Specificity , Tissue Distribution , Transfection
10.
Methods Mol Med ; 65: 61-78, 2001.
Article in English | MEDLINE | ID: mdl-21318745

ABSTRACT

Polycation-DNA complexes represent promising synthetic vectors for gene delivery, showing good transfection activities in vitro and safety in vivo. However, simple polycation-DNA complexes suffer from several disadvantages that limit their potential usefulness in vivo. Advances in this field thus rely on better control of the structure, colloidal, and surface properties of condensed DNA particles.

11.
Methods Mol Med ; 65: 131-48, 2001.
Article in English | MEDLINE | ID: mdl-21318751

ABSTRACT

There is an urgent requirement in the field of gene therapy for gene transfer vectors that are both safe to use and able to efficiently deliver therapeutic genes to target cells in vivo. Viral vectors, such as retrovirus, adenovirus, and herpes simplex virus, are efficient in transducing a broad range of cells, but they often lead to an inflammatory response against successfully transduced tissues, along with a strong immunogenicity of the virus itself (1). A further problem is the often expensive and laborious procedure required to produce the virus in sufficient quantities. In the past decade, several nonviral gene transfer vectors based on polycations (2) and liposomes (3,4), have been developed in order to overcome such problems. These vectors are becoming increasingly popular for use in delivering DNA to target cells both in vitro and in vivo because they are generally nonimmunogenic and easier to manufacture in bulk quantities. This chapter focuses on the use of polycations in gene delivery vectors.

12.
Biochim Biophys Acta ; 1475(3): 245-55, 2000 Jul 26.
Article in English | MEDLINE | ID: mdl-10913823

ABSTRACT

To investigate the possibility of producing charge-neutral gene delivery complexes with extended, non-particulate structures, DNA was allowed to self-assemble with a series of hydrophilic cationic polymers containing quaternary charged trimethylammonio ethylmethacrylate (TMAEM, 5, 15, 50, 100 mol%) copolymerised with hydrophilic N-(2-hydroxypropyl)methacrylamide (HPMA, 95, 85, 50, 0 mol%, respectively). Copolymers were all able to bind DNA, assessed using ethidium bromide fluorescence, although copolymers with low TMAEM content did not expel ethidium bromide. Increasing TMAEM content of the copolymers changed the morphology of the complexes from extended (5-15 mol% TMAEM), through partially condensed particles (50 mol%) to discrete nanoparticles (100 mol% TMAEM). Complexes based on copolymers with low TMAEM content (5-50 mol%) showed less resistance to degradation by nucleases and lower surface charge (21.2+/-5.9-45.1+/-3.9 mV) than those formed using 100 mol% TMAEM (57.8+/-8.2 mV). They also showed significantly less association with phagocytic cells in vitro (human leucocytes, uptake decreased by up to 92.3%; murine peritoneal macrophages, uptake decreased by up to 69.6%), although in vivo their hepatic accumulation was only slightly decreased (maximum decrease 27.6%). Finding the appropriate balance of hydrophilicity and stability is key to development of effective vectors for gene delivery.


Subject(s)
DNA/chemistry , Genetic Therapy/methods , Polymers/chemistry , Animals , Cations/chemistry , DNA/administration & dosage , DNA/ultrastructure , Endonucleases , Ethidium , Gene Transfer Techniques , Humans , Leukocytes/physiology , Macrophages, Peritoneal/physiology , Methacrylates/analysis , Methacrylates/chemistry , Mice , Microscopy, Electron , Microscopy, Fluorescence , Phagocytosis , Polyamines/chemistry , Polyelectrolytes , Polymers/administration & dosage , Static Electricity
13.
Eur J Pharm Sci ; 10(3): 169-77, 2000 May.
Article in English | MEDLINE | ID: mdl-10767594

ABSTRACT

The aim of this study was to evaluate the use of cationic-hydrophilic copolymers for self-assembly with antisense oligonucleotides targeted to the bcl-2 mRNA in order to improve their biocompatibility and modulation of their pharmacokinetics for greater therapeutic usefulness. Examination of the ability of poly(trimethylammonioethyl methacrylate chloride)-poly[N-(2-hydroxypropyl)methacrylamide] (pHPMA-b-pTMAEM) block copolymers to condense the oligonucleotide by fluorescence and electrophoresis techniques showed that complexes were formed more efficiently than with copolymers containing poly(ethylene glycol) blocks grafted onto the backbone of poly(L-lysine) (pLL-g-pEG). In addition, the copolymer pTMAEM-b-pHPMA produced oligonucleotide complexes with the most favourable physicochemical properties appropriate for in vivo applications. The complexes were small (approximately 36 nm in diameter), with low surface charge as measured by zeta potential, relatively stable to physiological salt conditions and could be formed at a DNA concentration of 500 microg/ml. Complex formation with the copolymer pTMAEM-b-pHPMA or pLL-g-pEG reduced the urinary clearance of the oligonucleotide after intravenous injection into mice. However after 30 min, the oligonucleotide complexes were cleared from the bloodstream. These results indicate that for the systemic delivery of oligonucleotides the polymer-derived complexes are not stable enough for prolonged circulation. Instead, these complexes may be more suitable for localised in vivo applications.


Subject(s)
Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/chemistry , RNA, Messenger/biosynthesis , RNA, Messenger/chemistry , Animals , Chemical Phenomena , Chemistry, Physical , DNA/chemistry , Electrophoresis, Agar Gel , Female , Intercalating Agents , Methacrylates , Mice , Mice, Inbred BALB C , Oligonucleotides, Antisense/pharmacokinetics , Particle Size , Pharmaceutical Vehicles , Polyethylene Glycols/chemistry , Polymers , Propidium , RNA, Messenger/pharmacokinetics , Spectrometry, Fluorescence , Surface Properties , Tissue Distribution
14.
J Biol Chem ; 275(6): 3793-802, 2000 Feb 11.
Article in English | MEDLINE | ID: mdl-10660529

ABSTRACT

Binding of serum proteins to polyelectrolyte gene delivery complexes is thought to be an important factor limiting bloodstream circulation and restricting access to target tissues. Protein binding can also inhibit transfection activity in vitro. In this study a multivalent reactive hydrophilic polymer has been used to inhibit protein binding. This polymer is based on poly-[N-(2-hydroxypropyl)methacrylamide] (pHPMA) bearing pendent oligopeptide (Gly-Phe-Leu-Gly) side chains terminated in reactive 4-nitrophenoxy groups (8.6 mol%). The polymer reacts with the primary amino groups of poly(L-lysine) (pLL) and produces a hydrophilic coating on the surface of pLL.DNA complexes (as measured by fluorescamine). The resulting pHPMA-coated complexes show a decreased surface charge (from +14 mV for pLL.DNA complexes to -25 mV for pHPMA-modified complexes) as measured by zeta potential analysis. The pHPMA-coated complexes also show a slightly increased average diameter (approximately 90 nm compared with 60 nm for pLL. DNA complexes) as viewed by atomic force and transmission electron microscopy and around 100 nm as viewed by photon correlation spectroscopy. They are completely resistant to protein interaction, as determined by turbidometry and SDS-polyacrylamide gel electrophoresis analysis of complexes isolated from plasma, and show significantly decreased nonspecific uptake into cells in vitro. Spare reactive ester groups can be used to conjugate targeting ligands (e.g. transferrin) on to the surface of the complex to provide a means of tissue-specific targeting and transfection. The properties of these complexes therefore make them promising candidates for targeted gene delivery, both in vitro and potentially in vivo.


Subject(s)
Gene Targeting/methods , Gene Transfer Techniques , Genetic Vectors/genetics , Polymers/chemistry , Transferrin/metabolism , Acrylic Resins/chemistry , Fluorescamine , Humans , Microscopy, Electron , Nephelometry and Turbidimetry , Particle Size , Peptides/chemistry , Polylysine/metabolism , Protein Binding , Serum Albumin/metabolism , Spectrum Analysis , Static Electricity , Surface Properties , Transfection , Tumor Cells, Cultured
15.
FEBS Lett ; 461(1-2): 96-100, 1999 Nov 12.
Article in English | MEDLINE | ID: mdl-10561503

ABSTRACT

A major factor limiting the development of non-viral gene delivery systems is the poor characterisation of polyelectrolyte complexes formed between cationic polymers and DNA. The present study uses the fluorescamine reagent to improve characterisation of poly(L-lysine) (pLL)/DNA complexes post-modified with a multivalent hydrophilic polymer by determining the availability of free amino groups. The results show that the fluorescamine reagent can be used to monitor the self-assembly reaction between pLL and DNA and the degree of surface modification of the resultant complexes with a hydrophilic polymer. This experimental approach should enable the preparation of fully defined complexes whose properties can be better related to their biological activity.


Subject(s)
DNA/metabolism , Fluorescamine/metabolism , Genetic Therapy/methods , Genetic Vectors/chemistry , Polylysine/metabolism , Animals , Cattle , Dose-Response Relationship, Drug , Ethidium/metabolism , Fluorescent Dyes/metabolism , Hydrogen-Ion Concentration , Kinetics , Molecular Weight , Nucleic Acid Conformation , Polymers/chemical synthesis , Thymus Gland/metabolism , Time Factors
16.
Gene Ther ; 6(4): 643-50, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10476224

ABSTRACT

Self-assembling polycation/DNA complexes represent a promising synthetic vector for gene delivery. However, despite considerable versatility and transfectional activity in vitro, such materials are quickly eliminated from the bloodstream following intravenous injection (plasma alpha half-life typically less than 5 min). For targeted systemic delivery a more prolonged plasma circulation of the vector is essential. Here we have examined factors contributing to rapid elimination of poly(L-lysine) (pLL)/DNA complexes from the bloodstream, and implicate the binding of proteins to the polyelectrolyte complexes as a likely cause for their blood clearance. pLL/DNA complexes reisolated from serum associate with several proteins, depending on their net charge, although the major band on SDS-PAGE co-migrates with albumin. Serum albumin binds to pLL/DNA complexes in vitro, forming a ternary pLL/DNA/albumin complex which regains some ethidium bromide fluorescence and fails to move during agarose electrophoresis. Albumin also causes increased turbidity of complexes, and reduces their zeta potential to the same level (-16 mV) as is measured in serum. We propose that rapid plasma elimination of polycation/DNA complexes results from their binding serum albumin and other proteins, perhaps due to aggregation and phagocytic capture or accumulation of the ternary complexes in fine capillary beds.


Subject(s)
DNA/metabolism , Genetic Therapy , Genetic Vectors/metabolism , Polylysine/metabolism , Transfection , Albumins/metabolism , Animals , Electrophoresis, Agar Gel , Female , Metabolic Clearance Rate , Mice , Mice, Inbred BALB C , Protein Binding , Treatment Failure
17.
FEBS Lett ; 418(1-2): 68-72, 1997 Nov 24.
Article in English | MEDLINE | ID: mdl-9414097

ABSTRACT

In the human insulin gene, a regulatory sequence upstream of the transcription start site at -229 to -258 (the E2 element) binds a ubiquitous factor USF. The present study led to the identification of a second factor, D0, that binds to an adjacent upstream site, the C2 element, that has previously not been described. The results demonstrate that D0 exhibits similar properties to RIPE3b1, a factor shown to be an important determinant of insulin gene beta-cell-specific expression. Binding of D0 to the C2 element was abolished by the oxidising agent diamide, and the alkylating agent N-ethylmaleimide. The results indicate that expression of the insulin gene may be regulated by a redox-dependent pathway involving RIPE3b1 or a RIPE3b1-like factor.


Subject(s)
DNA-Binding Proteins/metabolism , Insulin/genetics , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid , Animals , Base Sequence , Binding Sites , Humans , Insulin/biosynthesis , Islets of Langerhans/metabolism , Oxidation-Reduction , Rats , Sequence Alignment , Transcription, Genetic
18.
FEBS Lett ; 413(2): 304-8, 1997 Aug 18.
Article in English | MEDLINE | ID: mdl-9280302

ABSTRACT

Persistent hyperinsulinaemic hypoglycaemia of infancy (PHHI), or nesidioblastosis, is a rare disorder which may be familial or sporadic, and which is characterized by unregulated secretion of insulin and profound hypoglycaemia in the neonate. The defect has been linked in some patients to mutations in the sulphonyl urea receptor gene (SUR). The present study investigated potential defects in the regulation of the insulin gene by glucose in a beta-cell line (NES 2Y) derived from a patient with PHHI. The results show that the insulin promoter is unresponsive to glucose in PHHI, and that this defect can be attributed to impaired expression of the transcription factor IUF1. Because IUF1 is involved not only in linking glucose metabolism to the control of the insulin, but is also a major regulator of beta-cell differentiation during embryogenesis, we propose that impaired expression of IUF1 contributes to beta-cell dysfunction in PHHI by leading to abnormal beta-cell differentiation.


Subject(s)
DNA-Binding Proteins , Gene Expression Regulation/physiology , Homeodomain Proteins , Insulin/genetics , Islets of Langerhans/physiology , Pancreatic Diseases/genetics , Transcription Factors/genetics , Cell Line , DNA/metabolism , Gene Expression Regulation/drug effects , Glucose/pharmacology , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/analysis , Recombinant Fusion Proteins , Trans-Activators/genetics , Transcription Factors/metabolism , Transcription, Genetic , Upstream Stimulatory Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...