Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Biotechnol J ; 5(4): 385-92, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20235145

ABSTRACT

The Wnt canonical signaling pathway is essential for the early development of eukaryotic organisms and plays a key role in cell proliferation, differentiation, and oncogenesis. Moreover, the Wnt canonical signaling pathway contributes to the self-renewal of mouse hematopoietic stem cells (HSCs). Here, we demonstrate artificial activation of the Wnt canonical signaling pathway by beta-catenin protein transduction. Constitutively active beta-catenin protein was introduced into human embryonic kidney HEK-293 cells using a polyethylenimine (PEI) cationization method, or with the BioPORTER protein transduction reagent. We have previously shown that modification with PEI effectively causes proteins to be internalized by living mammalian cells. PEI-cationized, constitutively active beta-catenin protein was added to HEK-293 cells, and induction of several Wnt/beta-catenin target genes was detected by real-time PCR. However, using BioPORTER to introduce active beta-catenin did not activate the Wnt canonical signaling pathway. Introduction of eGFPNuc (enhanced green fluorescent protein variant containing a nuclear localization signal) into HEK-293 cells using the BioPORTER reagent caused significant cell death, as determined by propidium iodide staining. In contrast, the PEI-modified eGFPNuc did not impair survival of HEK-293 cells. These results indicate that the Wnt canonical signaling pathway could be successfully activated by transduction of PEI-cationized active beta-catenin, and the PEI-cationization method is an effective and safe technology for protein transduction into mammalian cells.


Subject(s)
Drug Carriers/chemistry , Kidney/physiology , Polyethyleneimine/chemistry , Signal Transduction/physiology , Transduction, Genetic/methods , Wnt Proteins/metabolism , beta Catenin/metabolism , Cations , Cell Line , Transfection/methods , beta Catenin/genetics
2.
Growth Factors ; 27(4): 228-36, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19521893

ABSTRACT

We found that eosinophil cationic protein (ECP) stimulated the growth of mouse Balb/c 3T3 fibroblasts. ECP-treated 3T3 cells were more flattened and exhibited enhanced stress fiber formation. The enhancement of cytoskeleton after addition of recombinant ECP appeared stable and was able to inhibit disassembly of actin filaments that was induced by fibroblast growth factor-2. The ROCK inhibitor, Y-27632, abrogated this enhancement on stress fiber formation that was induced by ECP indicating the involvement of Rho/ROCK signaling pathway. The effect of ECP was assessed on the differentiation of primary cardiomyocytes derived from rat neonatal heart since the development of actin filaments is significantly related with organization of stress fibers. As the result, both beating rate and the expression of cardiac muscle specific markers such as atrial natriuretic factor were enhanced in the presence of ECP. Thus ECP may also function as a cardiomyocyte differentiation factor.


Subject(s)
Eosinophil Cationic Protein/physiology , Myocytes, Cardiac/cytology , 3T3 Cells , Actins/metabolism , Amides/pharmacology , Animals , Cell Differentiation , Cytoskeleton/metabolism , Enzyme Inhibitors/pharmacology , Humans , Mice , Mice, Inbred BALB C , Myocytes, Cardiac/metabolism , Pyridines/pharmacology , Rats , Signal Transduction , Stress, Mechanical
3.
J Biochem ; 144(4): 447-55, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18603589

ABSTRACT

The glutathione S-transferase (GST)-fused protein expression system has been extensively used to generate a large quantity of proteins and has served for functional analysis in vitro. In this study, we developed a novel approach for the efficient intracellular delivery of GST-fused proteins into living cells to expand their usefulness up to in vivo use. Since protein cationization techniques are powerful strategies for efficient intracellular uptake by adsorptive-mediated endocytosis, GST-fused proteins were cationized by forming a complex with a polycationic polyethylenimine (PEI)-glutathione conjugate. On screening of protein transduction, optimized PEI-glutathione conjugate for protein transduction was characterized by a partly oligomerized mixture of PEI with average molecular masses of 600 (PEI600) modified with multiple glutathiones, which could have sufficient avidity for GST. Furthermore, enhanced endosomal escape of transduced GST-fused proteins was observed when they were delivered with a glutathione-conjugated PEI600 derivative possessing a hydroxybutenyl moiety. These results were confirmed by both intracellular confocal imaging of GST-fused green fluorescent protein and activation of an endogenous growth signal transduction pathway by a GST-fused constitutively active mutant of a kinase protein. These PEI-glutathione conjugates seem to be convenient molecular tools for protein transduction of widely used GST-fused proteins.


Subject(s)
Glutathione Transferase/metabolism , Glutathione/metabolism , Polyethyleneimine/metabolism , Animals , BALB 3T3 Cells , Biological Transport, Active , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , Cricetulus , Drug Carriers , Endocytosis , Glutathione Transferase/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , Mice , Molecular Weight , Polyethyleneimine/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transduction, Genetic
4.
J Biosci Bioeng ; 105(1): 34-8, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18295717

ABSTRACT

Polyethylenimine (PEI) cationization is a powerful strategy for protein transduction into cells. In this study, we attempted the artificial regulation of cell proliferation by protein transduction of the N-terminal domain (1-132 amino acids) of the simian virus 40 large T-antigen (SVLT-N), which inactivates retinoblastoma family proteins but not p53. To deliver SVLT-N into cells, we employed an indirect cationization method by forming a complex of biotynylated SVLT-N through disulfide bonds (biotin-SS-SVLT-N) and PEI-cationized avidin (PEI600-avidin). Using this complex, SVLT-N was transduced into the nucleus of confluent and quiescent Balb/c 3T3 cells and was found to be complexed with a cellular target protein, pRb. Furthermore, SVLT-N transduction induced cell proliferation in spite of confluent conditions. Because SVLT-N thus transduced into cells gradually degraded and was not detectable after a 4-d incubation, transiently transformed cells were obtained by this method. These results suggest that oncogene protein transduction technology has great potential for in vitro regulation of cell proliferation.


Subject(s)
Antigens, Polyomavirus Transforming/chemistry , Antigens, Polyomavirus Transforming/pharmacology , Avidin/chemistry , Cell Proliferation/drug effects , Polyethyleneimine/chemistry , Simian virus 40 , Animals , Antigens, Polyomavirus Transforming/metabolism , BALB 3T3 Cells , Biotinylation , Methods , Mice , Protein Structure, Tertiary , Protein Transport , Retinoblastoma Protein/metabolism
5.
Mol Biol Cell ; 19(1): 78-85, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17978094

ABSTRACT

We previously revealed a novel signal pathway involving S100A11 for inhibition of the growth of normal human keratinocytes (NHK) caused by high Ca(++) or transforming growth factor beta. Exposure to either agent resulted in transfer of S100A11 to nuclei, where it induced p21(WAF1). In contrast, S100A11 has been shown to be overexpressed in many human cancers. To address this apparent discrepancy, we analyzed possible new functions of S100A11, and we provide herein evidence that 1) S100A11 is actively secreted by NHK; 2) extracellular S100A11 acts on NHK to enhance the production of epidermal growth factor family proteins, resulting in growth stimulation; 3) receptor for advanced glycation end products, nuclear factor-kappaB, Akt, and cAMP response element-binding protein are involved in the S100A11-triggered signal transduction; and 4) production and secretion of S100A11 are markedly enhanced in human squamous cancer cells. These findings indicate that S100A11 plays a dual role in growth regulation of epithelial cells.


Subject(s)
Keratinocytes/cytology , S100 Proteins/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Epidermal Growth Factor/genetics , Humans , Keratinocytes/drug effects , Keratinocytes/enzymology , Keratinocytes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , S100 Proteins/pharmacology , Signal Transduction/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
6.
J Biol Chem ; 282(49): 35679-86, 2007 Dec 07.
Article in English | MEDLINE | ID: mdl-17932043

ABSTRACT

Regulation of cell growth and apoptosis is one of the pleiotropic functions of annexin A1 (ANXA1). Although previous reports on the overexpression of ANXA1 in many human cancers and on growth suppression and/or induction of apoptosis by ANXA1 may indicate the tumor-suppressive nature of ANXA1, molecular mechanisms of the function of ANXA1 remain largely unknown. Here we provide evidence that ANXA1 mechanistically links the epidermal growth factor-triggered growth signal pathway with cytosolic phospholipase A(2) (cPLA(2)), an initiator enzyme of the arachidonic acid cascade, through interaction with S100A11 in normal human keratinocytes (NHK). Ca(2+)-dependent binding of S100A11 to ANXA1 facilitated the binding of the latter to cPLA(2), resulting in inhibition of cPLA(2) activity, which is essential for the growth of NHK. On exposure of NHK to epidermal growth factor, ANXA1 was cleaved solely at Trp(12), and this cleavage was executed by cathepsin D. In squamous cancer cells, this pathway was shown to be constitutively activated. The newly found mechanistic intersection may be a promising target for establishing new measures against human cancer and other cell growth disorders.


Subject(s)
Annexin A1/metabolism , Carcinoma, Squamous Cell/metabolism , Keratinocytes/metabolism , Phospholipases A2, Cytosolic/metabolism , S100 Proteins/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Annexin A1/genetics , Apoptosis/genetics , Calcium/metabolism , Carcinoma, Squamous Cell/genetics , Cathepsin D/genetics , Cathepsin D/metabolism , Cell Line, Tumor , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Humans , Phospholipases A2, Cytosolic/genetics , Protein Binding/genetics , S100 Proteins/genetics , Signal Transduction/genetics , Tumor Suppressor Proteins/genetics
7.
Expert Opin Drug Discov ; 2(2): 261-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-23496081

ABSTRACT

The development of a method for the efficient intracellular delivery of inherently non-permeable proteins is needed for manipulation of cellular phenotypes or the discovery of protein-based drugs. It has been demonstrated that proteins artificially cationized by chemical conjugation show efficient intracellular delivery via adsorptive-mediated endocytosis and then can exert their biological activity in cells. Studies have also revealed that cationic peptides known as cell-penetrating peptides (CPPs) provide a means to deliver molecules into mammalian cells. Although the internalization mechanisms remain controversial, it is now becoming clear that the main port of entry into cells by CPPs also involves adsorptive-mediated endocytosis rather than the direct penetration of the plasma membrane. As the mammalian cell membrane possesses an abundance of negatively charged glycoproteins and glycosphingolipids, cationization of proteins is a reasonable choice to endow them with the ability for intracellular delivery. Cationization of proteins is usually accompanied by drastic changes in protein properties, structure and biological activities. Recently developed sophisticated protein chemistry can minimize these side effects. Therefore, protein cationization techniques will hopefully prove to be powerful tools for innovative research and drug discovery. In this review, techniques for cationization of proteins and their intracellular delivery, as well as some of their potential therapeutic applications, are discussed.

8.
Biochemistry ; 45(19): 6124-32, 2006 May 16.
Article in English | MEDLINE | ID: mdl-16681385

ABSTRACT

Cationization is a powerful strategy for internalizing a protein into living cells. On the other hand, a reversibly cationized denatured protein through disulfide bonds is not only soluble in water but also able to fold to the native conformation in vitro. When these advantages in cationization were combined, we developed a novel method to deliver a denatured protein into cells and simultaneously let it fold to express its function within cells. This "in-cell folding" method enhances the utility of recombinant proteins expressed in Escherichia coli as inclusion bodies; that is, the recombinant proteins in inclusion bodies are solubilized by reversible cationization through cysteine residues by disulfide bonds with aminopropyl methanethiosulfonate or pyridyldithiopropionylpolyethylenimine and then incubated with cells without an in vitro folding procedure. As a model protein, we investigated human tumor-suppressor p53. Treatment of p53-null Saos-2 cells with reversibly cationized p53 revealed that all events examined as indications of the activation of p53 in cells, such as reduction of disulfide bonds followed by tetramer formation, localization into the nucleus, induction of p53 target genes, and induction of apoptosis of cells, occurred. These results suggest that reversible cationization of a denatured protein through cysteine residues is an alternative method for delivery of a functional protein into cells. This method would be very useful when a native folded protein is not readily available.


Subject(s)
Tumor Suppressor Protein p53/metabolism , Base Sequence , Blotting, Western , Cations , DNA/metabolism , DNA Probes , Electrophoretic Mobility Shift Assay , Immunoprecipitation , Protein Binding , Protein Denaturation , Protein Folding , Protein Structure, Quaternary , Protein Transport , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tumor Suppressor Protein p53/chemistry
9.
J Biosci Bioeng ; 99(2): 95-103, 2005 Feb.
Article in English | MEDLINE | ID: mdl-16233763

ABSTRACT

In the post-genomic era, there is pressing need for development of protein manipulation methodology to analyze functions of proteins in living cells. For this purpose, techniques to deliver functional proteins into living cells are currently being evaluated as alternative approaches to the introduction of transcriptionally active DNA. Here, we describe a novel method for efficient protein transduction into living cells in which a protein is simply cationized with polyethylenimine (PEI) by limited chemical conjugation. PEI-cationized proteins appear to adhere to the cell surface by ionic charge interaction and then internalize into cells in a receptor- and transporter-independent fashion. Since PEI is an organic macromolecule with a high cationic-charge density, limited coupling with PEI results in endowment of sufficient cationic charge to proteins without causing serious decline in their fundamental functions. A number of PEI-cationized proteins, such as ribonuclease (RNase), green fluorescent protein (GFP) and immunoglobulin (IgG), efficiently entered cells and functioned in the cytosol. Our results suggest that protein cationization techniques using PEI will be useful for the development of protein transduction technology.


Subject(s)
Drug Delivery Systems/methods , Endocytosis/physiology , Intracellular Fluid/chemistry , Intracellular Fluid/metabolism , Polyethyleneimine/chemistry , Proteins/chemistry , Proteins/pharmacokinetics , 3T3 Cells , Animals , Cations , Diffusion , Injections, Intraperitoneal , Kidney/chemistry , Kidney/metabolism , Liver/chemistry , Liver/metabolism , Metabolic Clearance Rate , Mice , Mice, Inbred C57BL , Organ Specificity , Protein Transport/physiology , Proteins/administration & dosage , Static Electricity
10.
J Biochem ; 137(6): 693-701, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16002991

ABSTRACT

Previously, we have reported that cationized-proteins covalently modified with polyethylenimine (PEI) (direct PEI-cationization) efficiently enter cells and function in the cytosol [Futami et al. (2005) J. Biosci. Bioeng. 99, 95-103]. However, it may be more convenient if a protein could be delivered into cells just by mixing the protein with a PEI-cationized carrier protein having a specific affinity (indirect PEI-cationization). Thus, we prepared PEI-cationized avidin (PEI-avidin), streptavidin (PEI-streptavidin), and protein G (PEI-protein G), and examined whether they could deliver biotinylated proteins and antibodies into living cells. PEI-avidin (and/or PEI-streptavidin) carried biotinylated GFPs into various mammalian cells very efficiently. A GFP variant containing a nuclear localization signal was found to arrive even in the nucleus. The addition of a biotinylated RNase A derivative mixed with PEI-streptavidin to a culture medium of 3T3-SV-40 cells resulted in remarkable cell growth inhibition, suggesting that the biotinylated RNase A derivative entered cells and digested intracellular RNA molecules. Furthermore, the addition of a fluorescein-labeled anti-S100C (beta-actin binding protein) antibody mixed with PEI-protein G to human fibroblasts resulted in the appearance of a fluorescence image of actin-like filamentous structures in the cells. These results indicate that indirect PEI-cationization using non-covalent interaction is as effective as the direct PEI-cationization for the transduction of proteins into living cells and for expression of their functions in the cytosol. Thus, PEI-cationized proteins having a specific affinity for certain molecules such as PEI-streptavidin, PEI-avidin and PEI-protein G are concluded to be widely applicable protein transduction carrier molecules.


Subject(s)
Biotechnology/methods , Polyethyleneimine/metabolism , Protein Transport , Animals , Avidin , BALB 3T3 Cells , Biotinylation , Carrier Proteins/metabolism , Cations/chemistry , Cations/metabolism , Cell Death , Fibroblasts , Humans , Mice , Microscopy, Confocal , Nerve Tissue Proteins , Polyethyleneimine/chemistry , Ribonuclease, Pancreatic , Streptavidin
11.
J Biochem ; 132(5): 737-42, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12417023

ABSTRACT

There have been some attempts to develop immunotoxins utilizing human RNase as a cytotoxic domain of antitumor agents. We have recently shown that only human RNase 3 (eosinophil cationic protein, ECP) among five human pancreatic-type RNases excels in binding to the cell surface and has a growth inhibition effect on several cancer cell lines, even though the RNase activity of RNase 3 is completely inhibited by the ubiquitously expressed cytosolic RNase inhibitor. This phenomenon may be explained by that RNase 3 is very stable against proteolytic degradation because RNase 3 internalized through endocytosis could have a longer life time in the cytosol, resulting in the accumulation of enough of it to exceed the concentration of RNase inhibitor, which allows the degradation of cytosolic RNA molecules. Thus, we compared the stabilities of human pancreatic-type RNases (RNases 1-5) and bovine RNase A by means of guanidium chloride-induced denaturation experiments based on the assumption of a two-state transition for unfolding. It was demonstrated that RNase 3 is extraordinarily stabler than either RNase A or the other human RNases (by more than 25 kJ/mol). Thus, our data suggest that in addition to its specific affinity for certain cancer cell lines, the stability of RNase 3 contributes to its unique cytotoxic effect and that it is important to stabilize a human RNase moiety through protein engineering for the design of human RNase-based immunotoxins.


Subject(s)
Blood Proteins/metabolism , Eosinophil Cationic Protein , Pancreas/enzymology , Ribonuclease, Pancreatic/metabolism , Ribonucleases , Animals , Blood Proteins/isolation & purification , Cattle , Enzyme Stability , Eosinophil Granule Proteins , Humans , Thermodynamics
12.
Eur J Biochem ; 269(1): 307-16, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11784325

ABSTRACT

Eosinophil cationic protein (ECP), one of the major components of basic granules of eosinophils, is cytotoxic to tracheal epithelium. However, the extent of this effect on other cell types has not been evaluated in vitro. In this study, we evaluated the effect of ECP on 13 mammalian cell lines. ECP inhibited the growth of several cell lines including those derived from carcinoma and leukemia in a dose-dependent manner. The IC(50) values on A431 cells, MDA-MB-453 cells, HL-60 cells and K562 cells were estimated to be approximately 1-5 microm. ECP significantly suppressed the size of colonies of A431 cells, and decreased K562 cells in G1/G0 phase. However, there was little evidence that ECP killed cells in either cell line. These effects of ECP were not enhanced by extending its N-terminus. Rhodamine B isothiocyanate-labeled ECP started to bind to A431 cells after 0.5 h and accumulated for up to 24 h, indicating that specific affinity for the cell surface may be important. The affinity of ECP for heparin was assessed and found to be reduced when tryptophan residues, one of which is located at a position in the catalytic subsite of ribonuclease in ECP, were modified. The growth-inhibitory effect was also attenuated by this modification. These results suggest that growth inhibition by ECP is dependent on cell type and is cytostatic.


Subject(s)
Blood Proteins/pharmacology , Growth Inhibitors/pharmacology , Ribonucleases , Animals , Blood Proteins/analysis , Cell Division/drug effects , Cells, Cultured , Eosinophil Granule Proteins , Humans , Rats
SELECTION OF CITATIONS
SEARCH DETAIL