Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Arch Toxicol ; 97(7): 1887-1897, 2023 07.
Article in English | MEDLINE | ID: mdl-37193757

ABSTRACT

We previously found that methylmercury induces expression of oncostatin M (OSM), which is released extracellularly and binds to tumor necrosis factor receptor 3 (TNFR3), possibly enhancing its own toxicity. However, the mechanism by which methylmercury causes OSM to bind to TNFR3 rather than to its known receptors, OSM receptor and LIFR, is unknown. In this study, we aimed to elucidate the effect of methylmercury modification of cysteine residues in OSM on binding to TNFR3. Immunostaining of TNFR3-V5-expressing cells suggested that methylmercury promoted binding of OSM to TNFR3 on the cell membrane. In an in vitro binding assay, OSM directly bound to the extracellular domain of TNFR3, and this binding was promoted by methylmercury. Additionally, the formation of a disulfide bond in the OSM molecule was essential for the binding of both proteins, and LC/MS analysis revealed that methylmercury directly modified the 105th cysteine residue (Cys105) in OSM. Next, mutant OSM, in which Cys105 was replaced by serine or methionine, increased the binding to TNFR3, and a similar effect was observed in immunoprecipitation using cultured cells. Furthermore, cell proliferation was inhibited by treatment with Cys105 mutant OSMs compared with wildtype OSM, and this effect was cancelled by TNFR3 knockdown. In conclusion, we revealed a novel mechanism of methylmercury toxicity, in which methylmercury directly modifies Cys105 in OSM, thereby inhibiting cell proliferation via promoting binding to TNFR3. This indicates a chemical disruption in the interaction between the ligand and the receptor is a part of methylmercury toxicity.


Subject(s)
Cysteine , Methylmercury Compounds , Oncostatin M/chemistry , Oncostatin M/metabolism , Methylmercury Compounds/toxicity , Receptors, Tumor Necrosis Factor , Cell Proliferation
2.
Sci Rep ; 11(1): 16174, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34376712

ABSTRACT

Oncostatin M (OSM) is a pleiotropic, interleukin-6 family inflammatory cytokine that plays an important role in inflammatory diseases, including inflammatory bowel disease, rheumatoid arthritis, and cancer progression and metastasis. Recently, elevated OSM levels have been found in the serum of COVID-19 patients in intensive care units. Multiple anti-OSM therapeutics have been investigated, but to date no OSM small molecule inhibitors are clinically available. To pursue a high-throughput screening and structure-based drug discovery strategy to design a small molecule inhibitor of OSM, milligram quantities of highly pure, bioactive OSM are required. Here, we developed a reliable protocol to produce highly pure unlabeled and isotope enriched OSM from E. coli for biochemical and NMR studies. High yields (ca. 10 mg/L culture) were obtained in rich and minimal defined media cultures. Purified OSM was characterized by mass spectrometry and circular dichroism. The bioactivity was confirmed by induction of OSM/OSM receptor signaling through STAT3 phosphorylation in human breast cancer cells. Optimized buffer conditions yielded 1H, 15N HSQC NMR spectra with intense, well-dispersed peaks. Titration of 15N OSM with a small molecule inhibitor showed chemical shift perturbations for several key residues with a binding affinity of 12.2 ± 3.9 µM. These results demonstrate the value of bioactive recombinant human OSM for NMR-based small molecule screening.


Subject(s)
Drug Discovery/methods , Oncostatin M/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Binding Sites , Cell Line, Tumor , Humans , Magnetic Resonance Spectroscopy/methods , Molecular Docking Simulation , Oncostatin M/chemistry , Oncostatin M/metabolism , Phosphorylation , Protein Binding , STAT3 Transcription Factor/metabolism , Small Molecule Libraries/chemistry
3.
Biochim Biophys Acta Gen Subj ; 1865(7): 129907, 2021 07.
Article in English | MEDLINE | ID: mdl-33845142

ABSTRACT

BACKGROUND: Oncostatin M (OSM) and leukemia inhibitory factor (LIF) are two important pro-inflammatory cytokines of the interleukin-6 (IL-6) family. The two cytokines mediated signaling was recently found to be closely associated with cancer and chronic inflammation, which represent promising therapeutic targets for the treatment of many solid tumors and inflammatory disease. As the most closely related members, cross-reactivity of them may result in undesired activation of off-target cells, leading to toxicity or lack of efficacy of the therapeutic effects. However, the mechanism of the cross-reactivity of OSM and LIF is not well understood. METHODS: In this work, protein-protein docking, molecular dynamics (MD) simulations with explicit solvent and post endpoints binding free energy (BFE) analysis were carried out to further understand the structural and energetic principles of interactions between the two cytokines and the shared receptor LIFR. RESULTS: For the first time, the simulation given a computational model of OSM-LIFR interaction, and provided significant insights into the mechanism of OSM and LIF cross-react with LIFR. The identified common features shared by OSM and LIF bind to LIFR involving 10 "conserved" residues (90% similarity) distributed at the binding site III comprised of AB loop, BC loop and D helix. In addition, 11 shared residues were identified in LIFR contribute 77.85% and 84.63% energies for OSM and LIF binding, which play a critical role in the formation of the two cytokine-receptor complexes. Moreover, the "nonconserved" residues at the same position of cytokines such as Asp41 in OSM and Pro51 in LIF as well as the three residues (Glu338, Asn201 and Glu260) in LIFR were also discovered. CONCLUSIONS: These important information may facilitate the rational design of novel chemical or biological agents with less toxicity and improved efficacy.


Subject(s)
Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism , Leukemia Inhibitory Factor/chemistry , Leukemia Inhibitory Factor/metabolism , Oncostatin M/chemistry , Oncostatin M/metabolism , Amino Acid Sequence , Binding Sites , Cross Reactions , Humans , Molecular Dynamics Simulation , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Sequence Homology
4.
J Biol Chem ; 293(18): 7017-7029, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29511087

ABSTRACT

Oncostatin M (OSM) and leukemia inhibitory factor (LIF) are closely related members of the interleukin-6 (IL-6) cytokine family. Both cytokines share a common origin and structure, and both interact through a specific region, termed binding site III, to activate a dimeric receptor complex formed by glycoprotein 130 (gp130) and LIF receptor (LIFR) in humans. However, only OSM activates the OSM receptor (OSMR)-gp130 complex. The molecular features that enable OSM to specifically activate the OSMR are currently unknown. To define specific sequence motifs within OSM that are critical for initiating signaling via OSMR, here we generated chimeric OSM-LIF cytokines and performed alanine-scanning experiments. Replacement of the OSM AB loop within OSM's binding site III with that of LIF abrogated OSMR activation, measured as STAT3 phosphorylation at Tyr-705, but did not compromise LIFR activation. Correspondingly, substitution of the AB loop and D-helix in LIF with their OSM counterparts was sufficient for OSMR activation. The alanine-scanning experiments revealed that residues Tyr-34, Gln-38, Gly-39, and Leu-45 (in the AB loop) and Pro-153 (in the D-helix) had specific roles in activating OSMR but not LIFR signaling, whereas Leu-40 and Cys-49 (in the AB loop), and Phe-160 and Lys-163 (in the D-helix) were required for activation of both receptors. Because most of the key amino acid residues identified here are conserved between LIF and OSM, we concluded that comparatively minor differences in a few amino acid residues within binding site III account for the differential biological effects of OSM and LIF.


Subject(s)
Oncostatin M Receptor beta Subunit/metabolism , Oncostatin M/metabolism , Amino Acids/chemistry , Amino Acids/metabolism , Binding Sites , Cytokine Receptor gp130/metabolism , Cytokines/metabolism , Humans , Leukemia Inhibitory Factor/metabolism , Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism , Mutagenesis, Site-Directed , Oncostatin M/chemistry , Oncostatin M/genetics , Oncostatin M Receptor beta Subunit/chemistry , Oncostatin M Receptor beta Subunit/genetics , Phosphorylation , Protein Binding , Receptors, OSM-LIF/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction
5.
J Atheroscler Thromb ; 25(4): 294-303, 2018 Apr 01.
Article in English | MEDLINE | ID: mdl-29238011

ABSTRACT

Plaque calcification develops by the inflammation-dependent mechanisms involved in progression and regression of atherosclerosis. Macrophages can undergo two distinct polarization states, that is, pro-inflammatory M1 phenotype in progression and anti-inflammatory M2 phenotype in regression. In plaque progression, predominant M1 macrophages promote the initial calcium deposition within the necrotic core of the lesions, called as microcalcification, through not only vesicle-mediated mineralization as the result of apoptosis of macrophages and vascular smooth muscle cells (VSMCs), but also VSMC differentiation into early phase osteoblasts. On the other hand, in plaque regression M2 macrophages are engaged in the healing response to plaque inflammation. In association with the resolution of chronic inflammation, M2 macrophages may facilitate macroscopic calcium deposition, called as macrocalcification, through induction of osteoblastic differentiation and maturation of VSMCs. Oncostatin M, which has been shown to promote osteoblast differentiation in bone, may play a pivotal role in the development of plaque calcification. Clinically, two types of plaque calcification have distinct implications. Macrocalcification leads to plaque stability, while microcalcification is more likely to be associated with plaque rupture. Statin therapy, which reduces cardiovascular mortality, has been shown to exert its dual actions on plaque morphology, that is, regression of atheroma and increment of macroscopic calcium deposits. Statins may facilitate the healing process against plaque inflammation by enhancing M2 polarization of macrophages. Vascular calcification has pleiotropic properties as pro-inflammatory "microcalcification" and anti-inflammatory "macrocalcification". The molecular mechanisms of this process in relation with plaque progression as well as plaque regression should be intensively elucidated.


Subject(s)
Plaque, Atherosclerotic/physiopathology , Vascular Calcification/physiopathology , Animals , Apoptosis , Cell Differentiation , Disease Progression , Endoplasmic Reticulum Stress , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Inflammation , Macrophages/metabolism , Muscle, Smooth, Vascular/metabolism , Oncostatin M/chemistry , Osteoblasts/metabolism , Phenotype , Positron-Emission Tomography
6.
Arch Biochem Biophys ; 604: 1-10, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27242325

ABSTRACT

In human osteoarthritic chondrocytes, the hyaluronan receptor CD44 undergoes proteolytic cleavage at the cell surface. CD44 cleavage is thought to require transit of CD44 into cholesterol-rich lipid rafts. The purpose of this study was to investigate whether statins exert a protective effect on articular chondrocytes due to diminution of cholesterol. Three model systems of chondrocytes were examined including human HCS-2/8 chondrosarcoma cells, human osteoarthritic chondrocytes and normal bovine articular chondrocytes. Treatment with IL-1ß + Oncostatin M resulted in a substantial increase in CD44 fragmentation in each of the three chondrocyte models. Pre-incubation with simvastatin prior to treatment with IL-1ß + Oncostatin M decreased the level of CD44 fragmentation, decreased the proportion of CD44 that transits into the lipid raft fractions, decreased ADAM10 activity and diminished the interaction between CD44 and ADAM10. In HCS-2/8 cells and bovine articular chondrocytes, fragmentation of CD44 was blocked by the knockdown of ADAM10. Inhibition of CD44 fragmentation by simvastatin also resulted in improved retention of pericellular matrix. Addition of cholesterol and farnesyl-pyrophosphate reversed the protective effects of simvastatin. Thus, the addition of simvastatin exerts positive effects on chondrocytes including reduced CD44 fragmentation and enhanced the retention of pericellular matrix.


Subject(s)
Cartilage, Articular/pathology , Chondrocytes/metabolism , Hyaluronan Receptors/metabolism , Simvastatin/chemistry , ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Cartilage, Articular/drug effects , Cattle , Cell Line, Tumor , Cell Membrane/metabolism , Cholesterol/chemistry , Chondrocytes/drug effects , Chondrosarcoma/metabolism , Gene Expression Regulation , Humans , Interleukin-1beta/metabolism , Membrane Microdomains/chemistry , Membrane Proteins/metabolism , Mevalonic Acid/chemistry , Oncostatin M/chemistry , Polyisoprenyl Phosphates/chemistry , RNA, Small Interfering/metabolism , Sesquiterpenes/chemistry
7.
J Phys Chem B ; 119(38): 12397-409, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26340667

ABSTRACT

High-resolution NMR and density functional theory (DFT) calculations have been applied to analysis of heparin pentasaccharide 3D structure in aqueous solution. The fully optimized molecular geometry of two pentasaccharide conformations (differing from each other in the form, one (1)C4 and the other (2)S0, of the sulfated iduronic acid residue) were obtained using the B3LYP/6-311+G(d,p) level of theory in the presence of solvent, the latter included as explicit water molecules. The presented approach enabled insight into variations of the bond lengths, bond angles, and torsion angles, formations of intra- and intermolecular hydrogen bonds, and ionic interactions in the two pentasaccharide conformations. A rather complex hydrogen bond network is formed, including inter-residue and intraresidue bonds between the NH group in the GlcN,3,6S with oxygens linked to C-2 at the IdoA2S residue and the glycosidic O-1 and the neighboring OSO3(-) group linked to C-3 in the same residue. On the other hand, because the first hydration shell is strongly influenced by strong ion-ion and ion-dipole interactions between sodium ions, sulfates, carboxylates, and -OH groups, ionic interactions play an important role in the stabilization of the 3D structure. The DFT-computed three-bond proton-proton coupling constants also showed that best agreement with experiment was obtained with a weighted average of 15:85 ((1)C4/(2)S0) of the sulfated iduronic acid forms indicating that the ratio is even more shifted toward the (2)S0 form than previously supposed. The DFT-computed pentasaccharide conformation differs from the previously published data, with the main changes at the glycosidic linkages, namely, the ψ1 torsion angles and the ϕ3 angle. The comparison of the glycosidic linkage torsion angle values in solution with the antithrombin-pentasaccharide complex also indicates that the pentasaccharide conformation changes upon binding to antithrombin III. The data supports the assumption that the protein selects the more populated (2)S0 conformer of heparin pentasaccharide and, consequently, the binding process of heparin pentasaccharide with antithrombin III is energetically more favorable than formerly expected.


Subject(s)
Heparin/chemistry , Oligosaccharides/chemistry , Oncostatin M/chemistry , Water/chemistry , Hydrogen Bonding , Magnetic Resonance Spectroscopy , Models, Chemical , Molecular Structure , Protons , Solutions/chemistry
8.
J Tissue Eng Regen Med ; 9(11): E124-34, 2015 Nov.
Article in English | MEDLINE | ID: mdl-23505217

ABSTRACT

In recent years, the advantages of menstrual blood-derived stem cells (MenSCs), such as minimal ethical considerations, easy access and high proliferative ability, have inspired scientists to investigate the potential of MenSCs in cell therapy of different diseases. In order to characterize the potency of these cells for future cell therapy of liver diseases, we examined the potential of MenSCs to differentiate into hepatocytes, using different protocols. First, the immunophenotyping properties and potential of MenSCs to differentiate into osteoblasts, adipocytes and chondrocytes were evaluated. Thereafter, the differentiation protocols developed by two concentrations of hepatocyte growth factor (HGF) and oncostatin M (OSM), in combination with other components in serum-supplemented or serum-free culture media, were also investigated. The sequential differentiation was monitored by real-time PCR, immunostaining and functional assays. Our primary data revealed that the isolated MenSCs exhibited mesenchymal stem cell markers in parallel to OCT-4 as an embryonic marker. Regardless of differentiation procedures, the developed cells expressed mature hepatocyte markers, such as albumin, tyrosine aminotransferase and cytokeratin-18 at the mRNA and protein levels. They also showed functional properties of hepatocytes, including albumin secretion, glycogen storage and cytochrome P450 7A1 expression. However, the degree of differentiation was dependent on the concentrations of HGF and OSM. Indeed, omission of serum during the differentiation process caused typical improvement in hepatocyte-specific functions. This study is a novel report demonstrating the differentiation potential of MenSCs into hepatocyte-like cells. We recommend a complementary serum-free differentiation protocol for enrichment of in vitro production of functional MenSC-derived hepatocyte-like cells that could lead to a major step toward applied stem cell therapy of chronic liver diseases.


Subject(s)
Hepatocytes/cytology , Menstruation/blood , Stem Cells/cytology , Adipocytes/cytology , Adult , Albumins/chemistry , Cell Differentiation , Cell Proliferation , Chondrocytes/cytology , Culture Media/chemistry , Culture Media, Serum-Free , Female , Gene Expression Regulation , Glycogen/chemistry , Hepatocyte Growth Factor/chemistry , Humans , Immunophenotyping , Keratin-18/chemistry , Liver/metabolism , Liver Diseases/therapy , Oncostatin M/chemistry , Osteoblasts/cytology , Phenotype , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction
9.
J Biol Chem ; 287(39): 32848-59, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22829597

ABSTRACT

Oncostatin M (OSM) and leukemia inhibitory factor are pleiotropic cytokines that belong to the interleukin-6 (IL-6) family. These cytokines play a crucial role in diverse biological events like inflammation, neuroprotection, hematopoiesis, metabolism, and development. The family is grouped together based on structural similarities and their ability to activate the transmembrane receptor glycoprotein 130 (gp130). The common structure among these cytokines defines the spacing and the orientation of binding sites for cell surface receptors. OSM is unique in this family as it can signal using heterodimers of gp130 with either leukemia inhibitory factor receptor (LIFR) (type I) or oncostatin M receptor (OSMR) (type II). We have identified a unique helical loop on OSM between its B and C helices that is not found on other IL-6 family cytokines. This loop is located near the "FXXK" motif in active site III, which is essential for OSM's binding to both LIFR and OSMR. In this study, we show that the BC loop does not play a role in OSM's unique ability to bind OSMR. Shortening of the loop enhanced OSM's interaction with OSMR and LIFR as shown by kinetic and equilibrium binding analysis, suggesting the loop may hinder receptor interactions. As a consequence of improved binding, these structurally modified OSMs exhibited enhanced biological activity, including suppressed proliferation of A375 melanoma cells.


Subject(s)
Leukemia Inhibitory Factor Receptor alpha Subunit/chemistry , Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism , Oncostatin M Receptor beta Subunit/chemistry , Oncostatin M Receptor beta Subunit/metabolism , Oncostatin M/chemistry , Oncostatin M/immunology , Amino Acid Motifs , Cell Line, Tumor , Humans , Kinetics , Leukemia Inhibitory Factor Receptor alpha Subunit/genetics , Oncostatin M/genetics , Oncostatin M Receptor beta Subunit/genetics , Protein Binding , Protein Structure, Tertiary
10.
J Biol Chem ; 284(42): 28543-53, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19700767

ABSTRACT

The small leucine-rich repeat proteins, fibromodulin and osteoadherin, have N-terminal extensions with a variable number of O-sulfated tyrosine residues. This modification combined with a number of aspartic and glutamic acid residues results in a highly negatively charged domain of less than 30 amino acids. We hypothesized that this domain shares functional properties with heparin regarding binding to proteins and polypeptides containing clusters of basic amino acids. Two other family members, PRELP and chondroadherin, have distinctly different clusters of basic amino acids in their N and C termini, respectively, and PRELP is known to bind to heparin via this domain. Another heparin-binding protein is the cytokine Oncostatin M, with a different cluster of basic amino acids in its C terminus. We used polypeptides representing these basic domains in solid phase assays and demonstrate interactions with the negatively charged N-terminal domain of fibromodulin and full-length osteoadherin. The tyrosine sulfate domains also bound heparin-binding proteins such as basic fibroblast growth factor-2, thrombospondin I, MMP13, the NC4 domain of collagen IX, and interleukin-10. Fibronectin with large heparin-binding domains did not bind, neither did CILP containing a heparin-binding thrombospondin type I motif without clustered basic amino acids. Affinity depends on the number and position of the sulfated tyrosine residues shown by different binding properties of 10-kDa fragments subfractionated by ion-exchange chromatography. These interactions may sequester growth factors, cytokines, and matrix metalloproteinases in the extracellular matrix as well as contribute to its organization.


Subject(s)
Antimicrobial Cationic Peptides/chemistry , Blood Proteins/chemistry , Carrier Proteins/chemistry , Extracellular Matrix Proteins/chemistry , Proteoglycans/chemistry , Tyrosine/chemistry , Amino Acid Motifs , Amino Acids/chemistry , Animals , Cattle , Collagen/chemistry , Fibromodulin , Humans , Oncostatin M/chemistry , Peptides/chemistry , Protein Binding , Protein Structure, Tertiary , Sulfates/chemistry
11.
Int J Mol Med ; 23(2): 161-72, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19148539

ABSTRACT

The binding of oncostatin M (OM) to type I and type II receptor complexes elicits various biological responses by activating MEK/ERK and JAK/STAT signaling pathways. Some OM effects are clinically desirable such as reducing hyperlipidemia through the activation of hepatic LDL receptor transcription, a downstream event of ERK activation. The OM pro-inflammatory responses via induction of acute phase protein gene expression have been associated with STAT activation. In this study, by conducting site-directed mutagenesis, bioassays and molecular modeling we have defined 4 OM residues that are differently involved in the activation of ERK or STAT signaling pathway in HepG2 cells. We show that mutation of Lys163 to alanine totally abolished OM-mediated signaling, possibly because such mutation causes the disruption of a stabilizing H-bond pattern at the OM interface with receptors. G120A mutation equally impaired activations of ERK and STAT signaling pathways also by impairing the OM/cognate protein interactions. Interestingly, mutations of Gln20 and Asn123 differentially affected OM signaling through the two pathways. Q20A and N123A retained strong activity in inducing ERK phosphorylation but they showed diminished ability in activating STAT1 and STAT3. We further showed that mutations at Gln20 and Asn123 reduced OM induction of inflammatory gene fibrinogen-beta to a greater extent than that of LDL receptor gene. The mutation of Asn123 is directly related to local structural modification at site 3 of OM. Collectively these results provide a structural basis of OM-mediated signaling and suggest a potential to improve OM therapeutic properties via structural modification.


Subject(s)
Mutation , Oncostatin M/genetics , Signal Transduction/genetics , Animals , COS Cells , Cell Line, Tumor , Cells, Cultured , Chlorocebus aethiops , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Models, Molecular , Mutagenesis, Site-Directed , Oncostatin M/chemistry , Oncostatin M/metabolism , RNA, Messenger/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Structure-Activity Relationship , Transfection
12.
Biotechnol Prog ; 24(5): 1132-41, 2008.
Article in English | MEDLINE | ID: mdl-19194923

ABSTRACT

The development of implantable engineered liver tissue constructs and ex vivo hepatocyte-based therapeutic devices are limited by an inadequate hepatocyte cell source. In our previous studies, embryoid body (EB)-mediated stem cell differentiation spontaneously yielded populations of hepatocyte lineage cells expressing mature hepatocyte markers such as albumin (ALB) and cytokeratin-18 (CK18). However, these cultures neither yielded a homogenous hepatocyte lineage population nor exhibited detoxification function typical of a more mature hepatocyte lineage cell. In this study, secondary culture configurations were used to study the effects of collagen sandwich culture and oncostatin-M (OSM) or S-nitroso-N-acetylpenicillamine (SNAP) supplementation of EB-derived hepatocyte-lineage cell function. Quantitative immunofluorescence and secreted protein analyses were used to provide insights into the long-term maintenance and augmentation of existing functions. The results of these studies suggest that SNAP, independent of the collagen supplementation, maintained the highest levels of ALB expression, however, mature liver-specific CK18 was only expressed in the presence of gel sandwich culture supplemented with SNAP. In addition, albumin secretion and cytochrome P450 detoxification studies indicated that this condition was the best for the augmentation of hepatocyte-like function. Maintenance and augmentation of hepatocyte-like cells isolated from heterogeneous EB cell populations will be a critical step in generating large numbers of functional differentiated cells for therapeutic use.


Subject(s)
Collagen/metabolism , Embryonic Stem Cells/metabolism , Hepatocytes/metabolism , S-Nitroso-N-Acetylpenicillamine/metabolism , Cell Culture Techniques , Cells, Cultured , Collagen/chemistry , Culture Media/chemistry , Culture Media/metabolism , Cytochrome P-450 Enzyme System/metabolism , Embryonic Stem Cells/cytology , Hepatocytes/cytology , Humans , Oncostatin M/chemistry , Oncostatin M/metabolism , S-Nitroso-N-Acetylpenicillamine/chemistry
13.
Trends Biotechnol ; 25(2): 48-55, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17173994

ABSTRACT

In this Opinion article, we describe a nanotechnology-based approach to immobilize and orient proteins onto surfaces using atomic clusters prepared by physical methods. This is relevant to future protein biochips where dilute arrays of protein binding sites, each designed to immobilize no more than one protein molecule, would be ideal. In the case of a surface consisting of size-selected atomic gold clusters, proteins containing free cysteine residues can chemisorb directly to the bare cluster surface, thus effecting oriented immobilisation. The selection of atomic gold clusters in the size range 1-100 atoms (<3nm in diameter) is intended to ensure that, typically, only one protein can bind directly to the cluster surface. These nanoclusters of a smaller size scale than that of the protein present minimal contact between the gold and the protein, and hence imply a reduced risk of protein denaturing compared with gold films or extended surfaces.


Subject(s)
Binding Sites/physiology , Gold/chemistry , Nanotechnology/methods , Protein Array Analysis/methods , Protein Conformation , Proteins/chemistry , Cysteine/chemistry , Green Fluorescent Proteins/chemistry , Horseradish Peroxidase/chemistry , Microscopy, Atomic Force , Oncostatin M/chemistry , Protein Binding , Protein Denaturation , Proteins/isolation & purification
14.
J Biol Chem ; 281(48): 36673-82, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17028186

ABSTRACT

Activation of the signaling transduction pathways mediated by oncostatin M (OSM) requires the binding of the cytokine to either type I OSM receptor (leukemia inhibitory factor receptor/gp130) or to type II OSM receptor (OSMR/gp130). In the present work we have developed an enzyme-linked immunosorbent assay detecting a soluble form of OSMR (sOSMR) secreted by glioblastoma, hepatoma, and melanoma tumor cell lines. sOSMR was also present in sera of healthy individuals, with increased levels in multiple myeloma. Molecular cloning of a corresponding cDNA was carried out, and it encoded for a 70-kDa protein consisting of a half cytokine binding domain containing the canonical WSXWS motif, an immunoglobulin-like domain, and the first half of a second cytokine binding domain with cysteines in fixed positions. Analysis of the soluble receptor distribution revealed a preferential expression in lung, liver, pancreas, and placenta. sOSMR was able to bind OSM and interleukin-31 when associated to soluble gp130 or soluble interleukin-31R, respectively, and to neutralize both cytokine properties. We have also shown that OSM could positively regulate the synthesis of its own soluble receptor in tumor cells.


Subject(s)
Cytokine Receptor gp130/physiology , Interleukins/physiology , Oncostatin M/physiology , Receptors, OSM-LIF/physiology , Alternative Splicing , Amino Acid Motifs , Amino Acid Sequence , Base Sequence , Cell Line, Tumor , Cytokine Receptor gp130/chemistry , Glycoside Hydrolases/metabolism , Humans , Interleukins/chemistry , Molecular Sequence Data , Oncostatin M/chemistry , Protein Binding , Protein Structure, Tertiary , Receptors, OSM-LIF/chemistry , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...