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










Publication year range
2.
Phys Chem Chem Phys ; 20(23): 15807-15816, 2018 Jun 13.
Article in English | MEDLINE | ID: mdl-29845162

ABSTRACT

Hybrid cyclic α/ß-peptides, in which one or more ß-amino acids are incorporated into the backbone, are gaining increasing interest as potential therapeutics, thanks to their ability to achieve enhanced binding affinities for a biological target through pre-organization in solution. The in silico prediction of their three dimensional structure through strategies such as MD simulations would substantially advance the rational design process. However, whether the molecular mechanics force fields are accurate in sampling highly constrained cyclopeptides containing ß-amino acids remains to be verified. Here, we present a systematic assessment of the ability of 8 widely used force fields to reproduce 79 NMR observables (including chemical shifts and 3J scalar couplings) on five cyclic α/ß-peptides that contain the integrin recognition motif isoDGR. Most of the investigated force fields, which include force fields from AMBER, OPLS, CHARMM and GROMOS families, display very good agreement with experimental 3J(HN,Hα), suggesting that MD simulations could be an appropriate tool in the rational design of therapeutic cyclic α-peptides. However, for NMR observables directly related to ß-amino acids, we observed a poor agreement with experiments and a remarkable dependence of our evaluation on the choice of Karplus parameters. The force field weaknesses herein unveiled might constitute a source of inspiration for further force field optimization.


Subject(s)
Amino Acids/chemistry , Magnetic Resonance Spectroscopy , Peptides, Cyclic/chemistry , Drug Design , Magnetic Resonance Spectroscopy/standards , Molecular Dynamics Simulation , Protein Binding
3.
Ann N Y Acad Sci ; 971: 355-8, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12438151

ABSTRACT

We have found that chromogranin A (CgA), a protein released in circulation by neuroendocrine cells and neurons, prevents the vascular leakage induced by tumor necrosis factor (TNF) in a mouse model. Studies of the mechanism of action showed that CgA and its NH(2)-terminal fragments inhibit TNF-induced vascular permeability by preventing endothelial cytoskeleton rearrangements. We propose that neuronal/endocrine secretion of CgA could contribute to the regulation of endothelial barrier function and the protection of vessels against plasma leakage in inflammatory diseases.


Subject(s)
Chromogranins/physiology , Endothelium, Vascular/cytology , Animals , Cells, Cultured , Chromogranin A , Humans , Inflammation , Mice , Mice, Inbred BALB C , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism
4.
Anal Biochem ; 298(2): 181-8, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11700972

ABSTRACT

Tumor pretargeting with biotinylated antibody/avidin complexes improves the therapeutic index of systemically administered biotin-tumor necrosis factor (TNF) conjugates. Since the number of biotins in this conjugate is known to be critical for activity, we have characterized the structure of different biotin-TNF conjugates, prepared by reaction with d-biotinyl-6-aminocaproic acid N-hydroxysuccinimide ester and identified the biotinylation sites by trypsin digestion, reverse-phase chromatography, and electrospray mass spectrometry analyses. The results have shown that N-terminal valine is a preferential biotinylation site at pH 5.8, half of biotins being located on the alpha-amino group of this residue in a conjugate bearing one biotin/trimer (on average). Moreover, evidence has been obtained to suggest that the remaining part of biotins are linked to the epsilon-amino group of lysine 128, 112, and 65, while lysine 11, 90, and 98 were practically unmodified. No evidence of O-biotinylation of serine, threonine and tyrosine was obtained.


Subject(s)
Biotin/metabolism , Tumor Necrosis Factor-alpha/metabolism , Amino Acid Sequence , Animals , Binding Sites , Biotinylation , Cells, Cultured , Chromatography, High Pressure Liquid/methods , Fibroblasts/physiology , Humans , Mass Spectrometry/methods , Mice , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Trypsin/metabolism
5.
Am J Physiol Cell Physiol ; 281(4): C1173-9, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11546653

ABSTRACT

We have investigated the role of p55 and p75 tumor necrosis factor receptors 1 and 2 (TNFR1 and TNFR2, respectively) in TNF-induced alteration of endothelial permeability in vitro and in vivo. Stimulation of TNFR1 with an agonist antibody or a receptor-selective TNF mutein increased the flux of (125)I-albumin through endothelial cell monolayers. An antagonist anti-TNFR1 antibody, but not antagonist anti-TNFR2 antibodies, blocked the activity of TNF in vitro. Stimulation of TNFR1, but not TNFR2, induced cytoskeletal reorganization associated with increased permeability. SB-203580, a p38 mitogen-activated protein kinase inhibitor, blocked TNFR1-induced cytoskeletal reorganization and permeability. A selective mouse TNFR1 agonist and human TNF, which binds to murine TNFR1, increased the leakage of trypan blue-albumin from liver vessels in mice. These results indicate that stimulation of TNFR1 is necessary and sufficient to increase endothelial permeability in vitro and in vivo. However, an antagonist anti-murine TNFR2 antibody partially inhibited the effect of murine TNF on liver vessels, suggesting that TNFR2 also plays a role in the regulation of TNF-induced vascular permeability in vivo.


Subject(s)
Antigens, CD/metabolism , Endothelium, Vascular/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antibodies/pharmacology , Antigens, CD/immunology , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/physiology , Cells, Cultured , Cytoskeleton/metabolism , Endothelium, Vascular/cytology , Humans , Liver/blood supply , Liver/metabolism , Mice , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Stress Fibers/metabolism , Umbilical Veins/cytology
6.
Eur Cytokine Netw ; 11(4): 580-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11125300

ABSTRACT

TNF-induced caspase activation is critically involved in both apoptosis and protection from cell necrosis. We have investigated the roles of the p55- and p75-TNF receptors (TNFR1 and TNFR2) in the induction of mouse L-M cell death in the presence of a caspase inhibitor (zVAD-fmk) and a transcription inhibitor (actinomycin D), i.e. under conditions in which protective pathways requiring caspase activation and protein synthesis were blocked. Cytometric analysis after TNF treatment showed that apoptosis was inhibited, while necrosis was highly activated. In contrast, apoptosis was observed in cells treated with TNF and actinomycin D alone. Stimulation of TNFR1 was sufficient to induce either cell necrosis or apoptosis, even when we blocked endogenous TNF with an anti-murine TNF antibody. Experiments based on the use of receptor-agonist and antagonist antibodies also showed that TNFR2 contributes to cell necrosis and apoptosis. Simultaneous stimulation of TNFR2 and TNFR1 with specific agonists indicated that TNFR2 functionally cooperates with TNFR1 to potentiate the response indirectly, by inducing endogenous TNF cytotoxicity. Caspase inhibitors enhanced the cytotoxic effect of endogenous TNF, suggesting that TNFR2 modulation can regulate the global necrotic response to TNF. TNFR2 modulation could play an important role in determining the response to TNF in pathophysiological conditions characterized by caspase down-regulation and local TNF production.


Subject(s)
Antigens, CD/physiology , Caspase Inhibitors , Necrosis , Receptors, Tumor Necrosis Factor/physiology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Antibodies, Monoclonal/immunology , Cysteine Proteinase Inhibitors/pharmacology , Cytotoxicity Tests, Immunologic , Dactinomycin/pharmacology , Humans , Mice , Mice, Inbred C57BL , Models, Biological , Protein Synthesis Inhibitors/pharmacology , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
7.
Nat Biotechnol ; 18(11): 1185-90, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11062439

ABSTRACT

The clinical use of tumor necrosis factor alpha (TNF) as an anticancer drug is limited to local treatments because of its dose-limiting systemic toxicity. We show here that murine TNF fused with CNGRC peptide (NGR-TNF), an aminopeptidase N (CD13) ligand that targets activated blood vessels in tumors, is 12-15 times more efficient than murine TNF in decreasing the tumor burden in lymphoma and melanoma animal models, whereas its toxicity is similar. Similarly, human NGR-TNF induced stronger antitumor effects than human TNF, even with 30 times lower doses. Coadministration of murine NGR-TNF with a CNGRC peptide or an anti-CD13 antibody markedly decreased its antitumor effects. Tumor regression, induced by doses of murine NGR-TNF lower than the LD50, was accompanied by protective immunity. In contrast, no cure was induced by TNF at any dose. These results suggest that targeted delivery of TNF to CD13 may enhance its immunotherapeutic properties. Moreover, these findings reveal the potential of tumor homing peptides to generate a new class of recombinant cytokines that compared to immunocytokines have a simpler structure, could be easier to produce and are potentially less immunogenic.


Subject(s)
CD13 Antigens/metabolism , Neoplasms/therapy , Tumor Necrosis Factor-alpha/therapeutic use , Animals , CD13 Antigens/genetics , CD13 Antigens/therapeutic use , Cell Line , Cell Separation , Chromatography, Gel , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , HL-60 Cells , Humans , Ligands , Lymphoma/drug therapy , Mass Spectrometry , Melanoma, Experimental/drug therapy , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Neoplasms/drug therapy , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Time Factors , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , U937 Cells
8.
J Biol Chem ; 275(38): 29257-63, 2000 Sep 22.
Article in English | MEDLINE | ID: mdl-10875933

ABSTRACT

Previous studies showed that chromogranin A (CgA), a glycoprotein stored and co-released with various hormones by neuroendocrine cells and neurons, can modulate cell adhesion. We have investigated the structure-activity relationships of CgA using fibroblasts and coronary artery smooth muscle cells in adhesion assays. A recombinant CgA fragment 1-78 and a peptide 7-57 containing reduced and alkylated cysteines (Cys(17) and Cys(38)) induced cell adhesion after adsorption onto solid phases at 50-100 nm. Peptides lacking the disulfide loop region, including residues 47-68, 39-59, and 39-68, induced cell adhesion, either bound to solid phases at 200-400 nm or added to the liquid phase at 5-10 microm, whereas peptide 60-68 was inactive, suggesting that residues 47-57 are important for activity. The effect of CgA-(1-78) was blocked by anti-CgA antibodies against epitopes including residues Arg(53), His(54), and Leu(57). Substitutions of residues His(54), Gln(55), and Asn(56) with alanine decreased the cell adhesion activity of peptide 47-68. These results suggest that the region 47-57 (RILSILRHQNL) contains a cell adhesion site and that the disulfide bridge is not necessary for the proadhesive activity. The ability of soluble peptides to elicit proadhesive effects suggests an indirect mechanism. The high sequence conservation and accessibility to antibodies suggest that this region is important for the physiological role of CgA.


Subject(s)
Chromogranins/chemistry , Chromogranins/physiology , Fibroblasts , Muscle, Smooth, Vascular , Amino Acid Sequence , Binding Sites , Cell Adhesion , Cell Line , Chromogranin A , Humans , Molecular Sequence Data , Structure-Activity Relationship
9.
Cancer Res ; 59(12): 2917-23, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10383155

ABSTRACT

The clinical use of tumor necrosis factor alpha (TNF) as an anticancer drug is limited to local or locoregional administration because of dose-limiting systemic toxicity. We investigated in animal models whether the therapeutic index of systemically administered human or murine TNF can be increased by tumor pretargeting strategies based on the biotin-avidin system. Pretargeting of s.c. mouse WEHI-164 fibrosarcoma and RMA lymphoma genetically engineered to express the Thy 1.1 antigen on the cell membrane was achieved by i.p. injection of a biotinylated anti-Thy 1.1 antibody and avidin. This pretreatment increased the antitumor activity of systemically administered biotin-TNF conjugates by at least 5-fold. In contrast, pretargeting did not increase the toxicity of biotin-TNF, as judged by animal survival and weight loss after treatment. Ex vivo analysis of tumor cells 24 h after treatment showed that biotin-TNF persisted for several hours on the surface of pretargeted tumors, but not when avidin was omitted. The potentiation of the antitumor effects was related primarily to indirect mechanisms, involving a host-mediated response. The results indicate that tumor pretargeting improves the antitumor activity of TNF. Tumor pretargeting with avidin, which is currently used to increase the uptake of radioactive-labeled biotin in patients, could represent a new strategy for improving the therapeutic index of TNF.


Subject(s)
Antineoplastic Agents/therapeutic use , Avidin/therapeutic use , Lymphoma/drug therapy , Sarcoma, Experimental/drug therapy , Tumor Necrosis Factor-alpha/therapeutic use , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/toxicity , Biotinylation , Drug Delivery Systems , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , Recombinant Proteins/toxicity , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/toxicity
10.
Eur J Biochem ; 256(2): 453-60, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9760187

ABSTRACT

The mature hen avidin encoded by a synthetic cDNA was expressed in Escherichia coli in an insoluble form. After resolubilization, renaturation and purification, a recovery of about 20 mg/l cell culture was obtained. ELISA assays indicated no apparent differences in biotin binding between the natural and recombinant avidins. In addition, an acidic avidin mutant, bearing the substitutions Lys3-->Glu, Lys9--> Glu, Arg26-->Asp and Arg124-->Leu of four exposed basic residues, was produced. The protein, expressed and renatured as wild-type avidin, showed unaltered biotin-binding activity. The acidic pI (approximately 5.5) and lack of aggregation of the mutant allowed easy electrophoretic analysis under non-denaturing conditions of the protein alone and of its complexes with biotin, biotinylated transferrin or peroxidase. Analysis of the sera from sensitized subjects revealed that the avidin mutant has altered antigenicity. Both recombinant avidins were crystallized and the three-dimensional structures solved by molecular replacement and refined to 0.22 nm resolution. The three-dimensional structures of the two recombinant molecules, in the absence of biotin and of glycosylation, are fully comparable with those of the natural hen avidin previously reported.


Subject(s)
Avidin/chemistry , Escherichia coli/genetics , Recombinant Proteins/chemistry , Animals , Antibodies/blood , Antigens/immunology , Avidin/analysis , Avidin/genetics , Binding, Competitive , Biotin/metabolism , Biotinylation , Chickens , Crystallography, X-Ray , Gene Expression/genetics , Isoelectric Point , Mass Spectrometry , Models, Molecular , Mutation/genetics , Protein Binding/genetics , Protein Denaturation , Recombinant Proteins/immunology , Streptavidin/analysis
11.
Cancer Res ; 58(17): 3866-72, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9731496

ABSTRACT

We have recently described a new strategy for targeting biotinylated tumor necrosis factor-alpha (TNF-alpha) to tumors, based on pretargeting with biotinylated antibodies and avidin. Here, we have analyzed the structure-activity relationships of several biotin-TNF-alpha conjugates and studied the mechanism of their interaction with avidin and TNF-alpha receptors on tumor cells. The study has been carried out using an in vitro model based on human melanoma Colo 38 cells and monoclonal antibody 225, an antibody against the high molecular weight melanoma-associated antigen. Immunochemical and cytotoxicity studies showed that biotin-TNF-alpha but not TNF-alpha persists for several hours on the surface of cells pretargeted with biotin-monoclonal antibody 225 and avidin and triggers cytolytic effects. Studies on the mechanism of action showed that biotin-TNF-alpha trimers can slowly dissociate from targeted cells in a bioactive form, through trimer-monomer-trimer transitions. Structure-activity relationship studies showed that nonbiotinylated subunits must be present in the biotin-TNF-alpha trimers for efficient release of bioactive TNF-alpha. Colo 38 cells targeted with biotin-TNF-alpha were able to kill mouse L-M cells in coculture experiments, indicating that the released TNF-alpha can interact also with TNF-alpha receptors expressed by bystander cells. In conclusion, the targeting complex works as a system that slowly releases bioactive TNF-alpha in the microenvironment of the targeted cell. This opens up the possibility that cells other than those reached by the targeting antibody (e.g., endothelial cells and local cells of the immune system) can be affected in vivo.


Subject(s)
Avidin/metabolism , Biotin/therapeutic use , Neoplasms/therapy , Tumor Necrosis Factor-alpha/therapeutic use , Animals , Binding Sites , Biotin/pharmacology , Humans , Melanoma/therapy , Mice , Receptors, Tumor Necrosis Factor/metabolism , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
12.
Eur J Biochem ; 248(3): 692-9, 1997 Sep 15.
Article in English | MEDLINE | ID: mdl-9342219

ABSTRACT

Vasostatins (VS) are vasoinhibitory peptides derived from the N-terminal domain of chromogranin A, a secretory protein present in the electron-dense granules of many neuroendocrine cells. In this work we describe a method for the production in Escherichia coli of large amounts of recombinant vasostatins, corresponding to chromogranin A residues 1-78 (VS-1), and 1-115 (VS-2), and the use of these materials for structure characterisation. The masses of both products were close to the expected values, by SDS/PAGE and mass spectrometry analysis. However, their hydrodynamic behaviours in size-exclusion chromatography corresponded to that of proteins with a larger size. SDS/PAGE analysis of VS-1 and VS-2 after cross-linking with disuccinimidyl suberate indicated that both polypeptides form dimers. VS-2 was almost entirely dimeric at > 4 microM, but rapidly converted to monomer after dilution to 70 nM. The rapid dimer-monomer transition of VS-2 after dilution could be part of a mechanism for regulating its activity and localising its action. Immunological studies of VS-1 have shown that residues 37-70 constitute a highly antigenic region characterised by an abundance of linear epitopes efficiently mimicked by synthetic peptides. The recombinant products and the immunological reagents developed in this work could be valuable tools for further investigating the structure and the function of chromogranin A and its fragments.


Subject(s)
Chromogranins/chemistry , Antibodies, Monoclonal/immunology , Blotting, Western , Chromatography, High Pressure Liquid , Chromogranin A , Chromogranins/genetics , Chromogranins/immunology , Chromogranins/isolation & purification , Cloning, Molecular , Cross-Linking Reagents , Dimerization , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Escherichia coli/genetics , Humans , Mass Spectrometry , Peptides/chemistry , Peptides/immunology , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Succinimides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...