Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Curr Pharm Biotechnol ; 9(3): 215-25, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18673287

ABSTRACT

Rana pipiens oocytes contain two homologues of pancreatic ribonuclease A that are cytostatic and cytotoxic to human cancer cells. Extensively studied Onconase is in advanced Phase IIIb clinical trials against malignant mesothelioma, while Amphinase is a novel enzyme in pre-clinical development. Onconase is the smallest (104 amino acid residues) member of the ribonuclease A superfamily while Amphinase (114 residues) is the largest among amphibian ribonucleases. Both enzymes share the characteristic frog ribonucleases C-terminal disulfide bond but another signature of this group, the N-terminal pyroglutamate, an integral part of Onconase active site is not conserved in Amphinase. Although Onconase and Amphinase are weak catalysts their enzymatic activities are required for cytostatic and cytotoxic activity. While it was postulated that tRNA is the primary substrate of Onconase in vivo there is also extensive indirect evidence that suggests other RNA species, in particular micro RNAs, may actually be the critical target of these ribonucleases. The cytostatic effects of Onconase and Amphinase are manifested as cell arrest in the G(1) cell cycle phase. Apoptosis then follows involving activation of endonucleases(s), caspases, serine proteases and transglutaminase. Onconase was shown to be strongly synergistic when combined with numerous other antitumor modalities. Onconase and Amphinase are highly cationic molecules and their preferential toxicity towards cancer cells (having distinctly higher negative charge compared to normal cells) may depend on increased binding efficiency to the cell surface by electrostatic interactions.


Subject(s)
Antineoplastic Agents/pharmacology , Oocytes/enzymology , Rana pipiens , Ribonucleases/pharmacology , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Clinical Trials as Topic , Enzyme Stability , Humans , Models, Molecular , Molecular Sequence Data , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , RNA, Transfer/antagonists & inhibitors , Ribonucleases/chemistry , Ribonucleases/isolation & purification , Sequence Alignment
2.
Int J Oncol ; 31(3): 663-9, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17671695

ABSTRACT

Onconase (ONC), an antitumor ribonuclease from oocytes of a frog Rana pipiens, capable of inducing apoptosis in many cell lines is synergistic with several other anticancer drugs. Since cytotoxic effects of numerous drugs are modulated by reactive oxygen intermediates (ROI), we have studied effects of ONC on the intracellular level of oxidants in several normal cell types as well as tumor cell lines. It is demonstrated for the first time that ONC substantially decreases the content of ROI in all cell lines studied. This effect depends on the ribonucleolytic activity of the enzyme and is due to both, decreased rate of ROI generation and accelerated rate of their degradation. Onconase decreases the mitochondrial transmembrane potential and consequently, generation of ATP. Simultaneously the enzyme decreases the expression of an antiapoptotic protein Bcl-2, and upregulates the proapoptotic Bax protein. These finding are consistent with the enzyme propensity to induce apoptosis. The observed antioxidant activity of ONC may be an important element of its cytotoxicity towards cancer cells. The enzyme seems to exert its biological activities by interfering with the redox system of cellular regulation.


Subject(s)
Antineoplastic Agents/pharmacology , Ribonucleases/physiology , Animals , Antineoplastic Agents/chemistry , Apoptosis , Cell Line, Tumor , Humans , Jurkat Cells , Oxidants/metabolism , Oxidation-Reduction , Oxidative Stress , Rana pipiens , Reactive Oxygen Species , Ribonucleases/metabolism , Superoxide Dismutase/metabolism
3.
Int J Oncol ; 21(6): 1245-50, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12429974

ABSTRACT

Onconase (Onc) is an amphibian ribonuclease of the pancreatic RNase family that is cytostatic and cytotoxic to several tumor lines. It also shows anti-tumor activity in mouse tumor models and is currently in phase III clinical trials. In animal tests and clinical trials Onc shows lesser toxicity and fewer side effects compared to most chemotherapeutic drugs. Intriguingly, repeated infusions of this protein do not cause apparent immunological reactions in patients. The aim of the present study was to investigate sensitivity to Onc of human lymphocytes during their mitogenic stimulation in response to the polyvalent mitogen phytohemagglutinin (PHA), and in mixed allogeneic lymphocyte cultures. Unexpectedly, we observed that frequency of cells undergoing activation-induced apoptosis was markedly increased in all cultures containing Onc. Apoptosis was measured by flow cytometry using markers that detect activation of caspases, the in situ presence of DNA strand breaks, and loss of fragmented DNA ('sub-G1' cell subpopulation). The enhancement of frequency of activation-induced apoptosis (up to 244%) was observed at 4.2-83 nM Onc concentration, which is at least an order magnitude lower than its minimal concentration reported to affect proliferation or induce apoptosis of leukemic and solid tumor cell lines. The cell cycle progression of lymphocytes that responded to PHA mitogenically was not affected at 8.3 or 83 nM Onc concentration. Because activation-induced apoptosis is the key mechanism regulating several in vivo immunological functions including induction of tolerance, the observed effects of Onc may explain the apparent lack of immune reactions to this protein in treated patients. The propensity of Onc to potentiate the activation-induced apoptosis suggests that this drug may have clinical utility as immunomodulating agent, e.g., to suppress transplant rejection or treat autoimmune diseases.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Egg Proteins/pharmacology , Lymphocyte Activation/drug effects , Lymphocytes/pathology , Ribonucleases/pharmacology , Animals , Caspase Inhibitors , Caspases/metabolism , Cell Cycle/drug effects , Cell Division/drug effects , DNA Damage/drug effects , Enzyme Inhibitors/pharmacology , Flow Cytometry , Humans , In Situ Nick-End Labeling , Lymphocytes/enzymology , Phytohemagglutinins/pharmacology , Propidium/metabolism , Rana pipiens
4.
Proc Natl Acad Sci U S A ; 98(4): 1410-5, 2001 Feb 13.
Article in English | MEDLINE | ID: mdl-11171964

ABSTRACT

An additivity-based sequence to reactivity algorithm for the interaction of members of the Kazal family of protein inhibitors with six selected serine proteinases is described. Ten consensus variable contact positions in the inhibitor were identified, and the 19 possible variants at each of these positions were expressed. The free energies of interaction of these variants and the wild type were measured. For an additive system, this data set allows for the calculation of all possible sequences, subject to some restrictions. The algorithm was extensively tested. It is exceptionally fast so that all possible sequences can be predicted. The strongest, the most specific possible, and the least specific inhibitors were designed, and an evolutionary problem was solved.


Subject(s)
Algorithms , Ovomucin/metabolism , Serine Endopeptidases/metabolism , Trypsin Inhibitors/metabolism , Amino Acid Sequence , Animals , Bacterial Proteins , Binding Sites , Cattle , Chymotrypsin/metabolism , Humans , Leukocyte Elastase/metabolism , Molecular Sequence Data , Pancreatic Elastase/metabolism , Subtilisins/metabolism
5.
Cell Mol Life Sci ; 57(8-9): 1306-16, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11028920

ABSTRACT

A detailed analysis of the composition and properties of hydrophobic nuclei and microclusters has been carried out for onconase. Two main hydrophobic nuclei in the onconase structure were detected. Their composition and shape were found to be very similar to those of RNase A, in accordance with the predictions made. The nuclei in onconase are more compact, the side-chain atoms of residues included in the nuclei in onconase form more contacts with the environment than in RNase A. The hydrophobic nuclei should be considered as individual structural units along with elements of the secondary structure. Differences in composition and conformation of exposed loops between onconase and RNase A were found. The additional hydrophobic clusters attached to the nuclei in onconase might be involved in the fixation of an appropriate conformation of site(s) for manifestation of the biological activity of onconase. A comparison of amphibian representatives of the RNase A superfamily was also made. The results obtained suggest that the availability of nonpolar residues in established key positions of amino acid sequences determines the characteristic fold of homologous proteins and the structure of the active site cleft.


Subject(s)
Egg Proteins/chemistry , Egg Proteins/metabolism , Ribonucleases/chemistry , Ribonucleases/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cattle , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Folding , Protein Structure, Secondary , Ranidae , Ribonuclease, Pancreatic/chemistry , Sequence Alignment , Sequence Homology, Amino Acid
6.
Leukemia ; 12(8): 1241-8, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9697879

ABSTRACT

Onconase is a 12 kDa protein homologous to pancreatic RNase A isolated from amphibian oocytes which shows cytostatic and cytotoxic activity in vitro, inhibits growth of tumors in mice and is in phase III clinical trials. The present study was aimed to reveal mechanisms by which onconase perturbs the cell cycle progression. Human histiocytic lymphoma U937 cells were treated with onconase and expression of cyclins D3 and E, as well as of the cyclin-dependent kinase inhibitors (CKIs) p16INK4A, p21WAF1/CIP1 and p27KIP1 (all detected immunocytochemically) was measured by multiparameter flow cytometry, in relation to the cell cycle position. Also monitored was the status of phosphorylation of retinoblastoma protein (pRb) by a novel method utilizing mAb which specifically detects underphosphorylated pRb in individual cells. Cell incubation with 170 nM onconase for 24 h and longer led to their arrest in G1 which was accompanied by a decrease in expression of cyclin D3, no change in cyclin E, and enhanced expression of all three CKIs. pRb was underphosphorylated in the onconase arrested G1 cells but was phosphorylated in the cells that were still progressing through S and G2/M in the presence of onconase. The cytostatic effect of onconase thus appears to be mediated by downregulation of cyclin D3 combined with upregulation of p27KIP1, p16INK4A and p21WAF1/CIP1, the events which may prevent phosphorylation of pRb during G0/1 and result in cell arrest at the restriction point controlled by Cdk4/6 and D type cyclins.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle Proteins , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclins/metabolism , Egg Proteins/pharmacology , Microtubule-Associated Proteins/metabolism , Retinoblastoma Protein/metabolism , Ribonucleases/pharmacology , Tumor Suppressor Proteins , Animals , Cyclin D3 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Inhibitors/metabolism , G1 Phase , Genes, Tumor Suppressor , Humans , Lymphoma, Large B-Cell, Diffuse/metabolism , Mice , Phosphorylation , Tumor Cells, Cultured
7.
Int J Oncol ; 13(1): 11-6, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9625797

ABSTRACT

Onconase (ONC) a ribonuclease from amphibian oocytes is cytostatic and cytotoxic to many human tumor lines, shows in vivo antitumor activity in mouse tumor models and is in Phase III clinical trials. The mechanism of antitumor activity of ONC is presumed to be due to its internalization, degradation of intracellular RNA and suppression of protein synthesis. Since apoptosis triggered by TNF-alpha is known to be potentiated by inhibitors of protein synthesis, we have hypothesized that it also may be potentiated by ONC. Indeed, preincubation of U-937 or HL-60 leukemic cells with 0.17 microM ONC rendered them more sensitive to induction of apoptosis by TNF-alpha or antibody to CD95 (Fas). The mechanism by which ONC amplifies the effect of TNF-alpha may involve suppression of induction of the survival genes whose expression is triggered by activation of NFkB by this factor.


Subject(s)
Apoptosis/drug effects , Egg Proteins/metabolism , Ribonucleases/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antigens, CD/metabolism , Drug Synergism , Fas Ligand Protein , HL-60 Cells , Humans , Membrane Glycoproteins/pharmacology , Mice , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I , Tumor Cells, Cultured , fas Receptor/metabolism
8.
J Biol Chem ; 271(34): 20783-8, 1996 Aug 23.
Article in English | MEDLINE | ID: mdl-8702832

ABSTRACT

Ribonucleases appear to have physiologic roles in host defense against cancer, viruses, and other parasites. Previously it was shown that select ribonucleases added to cells concurrently with virions blocked human immunodeficiency virus, type I (HIV-1) infection of H9 cells. We now report that a ribonuclease homologous to RNase A, named onconase, inhibits virus replication in chronically HIV-1-infected human cells without killing the virally infected cell. Examining the mechanism of this inhibition shows that onconase enters the infected cells and degrades HIV-1 RNA without degrading ribosomal RNA or the three different cellular messenger RNAs analyzed. The homologous human pancreatic RNase lacks anti-viral activity. Comparing recombinant forms of onconase and a onconase-human RNase chimera shows that the N-terminal 9 amino acids and the pyroglutamyl residue of onconase are required for full anti-viral activity. Thus extracellular ribonucleases can enter cells, metabolize select RNAs, and inhibit HIV virion production within viable replicating cells.


Subject(s)
Antiviral Agents , Egg Proteins/metabolism , HIV-1/growth & development , RNA, Viral/metabolism , Ribonucleases/metabolism , Egg Proteins/pharmacology , Extracellular Space/enzymology , HIV Core Protein p24/analysis , Humans , RNA, Messenger/metabolism , RNA, Ribosomal/metabolism , Recombinant Proteins , Ribonucleases/pharmacology , Structure-Activity Relationship , Substrate Specificity , Tumor Cells, Cultured , Virus Replication/drug effects
9.
J Natl Cancer Inst ; 88(11): 747-53, 1996 Jun 05.
Article in English | MEDLINE | ID: mdl-8637029

ABSTRACT

BACKGROUND: Onconase, a protein isolated from oocytes and early embryos of the frog Rana pipiens, shares extensive homology with bovine pancreatic ribonuclease (RNase A) and possesses similar enzyme activity. Onconase is cytotoxic toward cancer cells in vitro and exhibits antitumor activity in animal models. In addition, Onconase has been shown to enhance the cytotoxic activity of some chemotherapeutic agents in vitro. PURPOSE: We studied interactions between the cytotoxic effects of Onconase and the chemotherapeutic agent vincristine (VCR) in the treatment of drug-sensitive and multidrug-resistant human colon carcinoma cells in vitro and in mice. METHODS: Transplantable human colon carcinoma cells (HT-29par cells) were infected with a retrovirus containing human mdr1 (also known as MDR1 and PGY1) complementary DNA (encoding P-glycoprotein [P-gp]), and clones that were cross-resistant to colchicine, doxorubicin, and vinblastine were selected (HT-29mdr1 cells). Drug-resistant HT-29mdr1 cells and drug-sensitive HT-29par parental cells were treated with Onconase and/or VCR in vitro at varying concentrations to measure the effects on protein synthesis and cell viability. The impact of Onconase on VCR accumulation in both types of cells was determined in the presence or absence of MRK-16, an anti-P-gp monoclonal antibody capable of reversing the multidrug-resistant phenotype. The antitumor effects of Onconase and/or VCR treatment were assessed in nude mice bearing established HT-29par or HT-29mdr1 intraperitoneal tumors. IC50 values (drug concentrations resulting in 50% inhibition of protein synthesis or cell viability) for Onconase and VCR were determined from semilogarithmic dose-response curves; interactions between the cytotoxic effects of these two agents were evaluated using data from protein synthesis inhibition experiments and a two-way analysis of variance. Survival distributions from in vivo experiments were compared using Cox proportional hazards models. RESULTS: The combination of Onconase and VCR yielded enhanced cytotoxicity in vitro that was independent of P-gp expression. Evaluation of the effects of these two compounds on protein synthesis over a wide range of drug concentrations indicated possible synergistic interactions (i.e., greater than additive effects) in both drug-resistant and drug-sensitive cells. The enhancement of VCR cytotoxicity was dependent on Onconase enzyme activity and was not associated with increased intracellular levels of VCR. Simultaneous treatment of mice bearing HT-29par tumors with Onconase and VCR did not extend their median survival time (MST) significantly (MST with VCR = 66 days; MST with VCR plus Onconase = 69 days; two-tailed P = .57); however, the MST of mice with HT-29mdr1 tumors was extended significantly by this treatment (MST with VCR = 44 days; MST with VCR plus Onconase = 66 days; two-tailed P<.001). CONCLUSION: Combined administration of Onconase and VCR yields enhanced cytotoxicity in vitro and in vivo against human colon carcinoma cells that overexpress the mdr1 gene.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Egg Proteins/pharmacology , Ribonucleases/pharmacology , Vincristine/pharmacology , Animals , Colonic Neoplasms/drug therapy , Drug Resistance , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Transplantation, Heterologous , Tumor Cells, Cultured , Vincristine/pharmacokinetics
10.
J Mol Biol ; 257(5): 992-1007, 1996 Apr 19.
Article in English | MEDLINE | ID: mdl-8632481

ABSTRACT

A number of biochemical properties differ dramatically among homologues within the pancreatic ribonuclease superfamily. Human pancreatic ribonuclease (hRNase) has high enzyme activity, extreme sensitivity to ribonuclease inhibitor (RI) and is non-toxic, whereas a homologous RNase from frog eggs, called onconase, has much lower enzyme activity, is not sensitive to RI and is cytotoxic to cancer cell lines and animals. To explore the structural basis of these differences among members in the RNAse family we synthesized genes for onconase, hRNase, a mutant onconase (K9Q) and onconase-hRNase N-terminal hybrids and expressed the proteins in Escherichia coli with final yields of 10 to 50 mg per liter of culture after purification. A recombinant version of onconase with an N-terminal methionine instead of the native pyroglutamyl residue had decreased cytotoxicity and enzyme activity. Cleavage of the recombinant onconase Met-1 residue, and cyclization of the Gln1 residue to reform the pyroglutamyl N terminus, reconstituted cytotoxicity and enzyme activity. Thus a unique role of the pyroglutamyl residue in the active site of amphibian RNases is indicated. Replacement of one to nine residues of onconase with the homologous residues of hRNase increased the enzymatic activity against most of the substrates tested with a simultaneous shift in the enzyme specificity from high preference for poly(U) to slight preference for poly(C). Cytotoxicity of the chimera decreased, dissociating cytotoxicity from enzymatic activity. The molecular basis for the low binding affinity of onconase for RI has been examined experimentally with the recombinant RNases and by fitting onconase and RNase A structures to the coordinates from the recently published RNase A-RI complex.


Subject(s)
Cell Survival/drug effects , Egg Proteins/metabolism , Enzyme Inhibitors/pharmacology , Placental Hormones/pharmacology , Ribonuclease, Pancreatic/metabolism , Ribonucleases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Egg Proteins/antagonists & inhibitors , Egg Proteins/chemistry , Egg Proteins/pharmacology , Humans , Kinetics , Molecular Sequence Data , Mutagenesis , Osmolar Concentration , Pyrrolidonecarboxylic Acid/chemistry , Pyrrolidonecarboxylic Acid/metabolism , RNA/metabolism , Rana pipiens , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Ribonuclease, Pancreatic/antagonists & inhibitors , Ribonuclease, Pancreatic/chemistry , Ribonuclease, Pancreatic/pharmacology , Ribonucleases/antagonists & inhibitors , Ribonucleases/chemistry , Ribonucleases/pharmacology , Sequence Homology, Amino Acid , Substrate Specificity , Tumor Cells, Cultured
11.
J Biol Chem ; 270(29): 17476-81, 1995 Jul 21.
Article in English | MEDLINE | ID: mdl-7542240

ABSTRACT

Several ribonucleases serve as cytotoxic agents in host defense and in physiological cell death pathways. Although certain members of the pancreatic ribonuclease A superfamily can be toxic when applied to the outside of cells, they become thousands of times more toxic when artificially introduced into the cytosol, indicating that internalization is the rate-limiting step for cytotoxicity. We have used three agents that disrupt the Golgi apparatus by distinct mechanisms, retinoic acid, brefeldin A, and monensin, to probe the intracellular pathways ribonucleases take to reach the cytosol. Retinoic acid and monensin potentiate the cytotoxicity of bovine seminal RNase, Onconase, angiogenin, and human ribonuclease A 100 times or more. Retinoic acid-mediated potentiation of ribonucleases is completely blocked by brefeldin A. Ribonucleases appear to route more efficiently into the cytosol through the Golgi apparatus disrupted by monensin or retinoic acid. Intracellular RNA degradation by BS-RNase increased more than 100 times in the presence of retinoic acid confirming that the RNase reaches the cytosol and indicating that degradation of RNA is the intracellular lesion causing toxicity. As retinoic acid alone and Onconase are in clinical trials for cancer therapy, combinations of RNases and retinoic acid in vivo may offer new clinical utility.


Subject(s)
Antineoplastic Agents/pharmacology , Ribonuclease, Pancreatic , Ribonucleases/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Brefeldin A , Cattle , Cell Survival/drug effects , Cyclopentanes/pharmacology , Egg Proteins/pharmacology , Golgi Apparatus/drug effects , Humans , Molecular Sequence Data , Monensin/pharmacology , Proteins/pharmacology , RNA/metabolism , Rats , Tretinoin/pharmacology , Tumor Cells, Cultured
12.
J Mol Biol ; 236(4): 1141-53, 1994 Mar 04.
Article in English | MEDLINE | ID: mdl-8120892

ABSTRACT

The X-ray crystallographic structure of P-30 protein (Onconase) has been solved by multiple isomorphous replacement and the structure has been refined at 1.7 A resolution to a conventional R-factor of 0.178. The molecular model comprises all 826 non-hydrogen protein atoms, 96 solvent molecules and a sulfate anion that is bound at the active site. The molecular structure is similar to that of ribonuclease A. The active site cleft is located at the junction of two three-stranded beta-sheets and the N-terminal helix. A sulfate anion is non-covalently bound by Lys9, His10, His97, Phe98 and an intermolecular contact involving Lys55' from a neighboring molecule. The N-terminal pyroglutamyl (Pyr) residue is part of the active site and its O epsilon 1 atom forms a hydrogen bond with the Lys9 N zeta. The previously constructed comparative molecular model of P-30 based on ribonuclease A correctly predicted the overall fold of P-30 and the conformation of its active site residues. The model failed to predict the conformation of Pyr1 and the conformation of the two loops following helix alpha 3 and strand beta 3.


Subject(s)
Antineoplastic Agents/chemistry , Egg Proteins/chemistry , Ribonucleases/chemistry , Animals , Binding Sites , Crystallography, X-Ray , Hydrogen Bonding , Models, Molecular , Molecular Structure , Protein Conformation , Rana pipiens
13.
J Neurosci ; 14(2): 538-44, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8301353

ABSTRACT

Purkinje cell toxicity is one of the characteristic features of the Gordon phenomenon, a syndrome manifested by ataxia, muscular rigidity, paralysis, and tremor that may lead to death (Gordon, 1933). Two members of the RNase superfamily found in humans, EDN (eosinophil-derived neurotoxin) and ECP (eosinophil cationic protein), cause the Gordon phenomenon when injected intraventricularly into guinea pigs or rabbits. We have found that another member of the RNase superfamily, an antitumor protein called onconase, isolated from Rana pipiens oocytes and early embryos, will also cause the Gordon phenomenon when injected into the cerebrospinal fluid of guinea pigs at a dose similar to that of EDN (LD50, 3-4 micrograms). Neurologic abnormalities of onconase-treated animals were indistinguishable from those of EDN-treated animals, and histology showed dramatic Purkinje cell loss in the brains of onconase-treated animals. The neurotoxic activity of onconase correlates with ribonuclease activity. Onconase modified by iodoacetic acid to eliminate 70% and 98% of the ribonuclease activity of the native enzyme displays a similar decrease in ability to cause the Gordon phenomenon. In contrast, the homologous bovine pancreatic RNase A injected intraventricularly at a dose 5000 times greater than the LD50 dose of EDN or onconase is not toxic and does not cause the Gordon phenomenon. A comparison of the RNase activities of EDN, onconase, and bovine pancreatic RNase A using three pancreatic RNA substrates demonstrates that onconase is orders of magnitude less active enzymatically than EDN and RNase A. Thus, another member of the RNase superfamily in addition to EDN and ECP can cause the Gordon phenomenon.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Antineoplastic Agents/toxicity , Blood Proteins/toxicity , Egg Proteins/toxicity , Neurotoxins/toxicity , Purkinje Cells/drug effects , Ribonuclease, Pancreatic/toxicity , Ribonucleases/toxicity , Amino Acid Sequence , Animals , Blood Proteins/administration & dosage , Blood Proteins/chemistry , Cerebral Ventricles/drug effects , Cerebral Ventricles/physiology , Egg Proteins/administration & dosage , Egg Proteins/chemistry , Eosinophil Granule Proteins , Eosinophil-Derived Neurotoxin , Female , Guinea Pigs , Humans , Injections, Intraventricular , Injections, Spinal , Molecular Sequence Data , Neurotoxins/administration & dosage , Neurotoxins/chemistry , Purkinje Cells/pathology , Rabbits , Ribonuclease, Pancreatic/administration & dosage , Ribonuclease, Pancreatic/chemistry , Ribonucleases/administration & dosage , Ribonucleases/chemistry , Sequence Homology, Amino Acid , Spinal Cord/drug effects , Spinal Cord/physiology
14.
J Biol Chem ; 268(14): 10686-93, 1993 May 15.
Article in English | MEDLINE | ID: mdl-8486718

ABSTRACT

Onconase, or P-30, is a protein initially purified from extracts of Rana pipiens oocytes and early embryos based upon its anticancer activity both in vitro and in vivo. It is a basic single-chain protein with an apparent molecular mass of 12,000 daltons and is homologous to RNase A. In cultured 9L glioma cells, onconase inhibits protein synthesis with an IC50 of about 10(-7) M. The inhibition of protein synthesis correlates with cell death determined by clonogenic assays. 125I-Labeled onconase binds to specific sites on cultured 9L glioma cells. Scatchard analysis of the binding data shows that onconase appears to bind to cells with two different affinities, one with a Kd of 6.2 x 10(-8) and another of 2.5 x 10(-7) M. Each cell could bind about 3 x 10(5) molecules of onconase at each of the two affinity sites. The low affinity Kd is similar to the IC50 for onconase toxicity. Onconase also demonstrates a saturability of cytotoxicity at a concentration that would saturate the low affinity binding site. Incubation at 4 degrees C increased the binding of onconase to cells relative to 37 degrees C binding and also increased the sensitivity of cells to onconase toxicity, indicating that receptor binding may be an initial step in cell toxicity. Onconase cytotoxicity can be blocked by metabolic inhibitors, NaN3 and 2-deoxyglucose, and cytotoxicity is potentiated 10-fold by monensin. Ribonuclease activity appears necessary for onconase toxicity because alkylated onconase, which only retains 2% of the ribonuclease activity, was at least 100-fold less potent in inhibiting protein synthesis in cells. Onconase inhibition of protein synthesis in 9L cells coincides with the degradation of cellular 28 S and 18 S rRNA. In contrast to RNase A, onconase is resistant to two RNase inhibitors, placental ribonuclease inhibitor and Inhibit-Ace. Northern hybridization with placental ribonuclease inhibitor cDNA probe indicates that 9L glioma cells contain endogenous placental ribonuclease inhibitor mRNA. Based on these results, we propose that onconase toxicity results from onconase binding to cell surface receptors, internalization to the cell cytosol where it degrades ribosomal RNA, inhibiting protein synthesis and causing cell death.


Subject(s)
Antineoplastic Agents/metabolism , Antineoplastic Agents/toxicity , Cell Survival/drug effects , Egg Proteins/metabolism , Egg Proteins/toxicity , Protein Synthesis Inhibitors/pharmacology , Ribonucleases/metabolism , Ribonucleases/toxicity , Animals , Autoradiography , Blotting, Northern , Brefeldin A , Cyclopentanes/pharmacology , Dose-Response Relationship, Drug , Embryo, Nonmammalian , Glioma , Kinetics , Leucine/metabolism , Neoplasm Proteins/biosynthesis , Oocytes , Placental Hormones/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rana pipiens , Ribonuclease, Pancreatic/metabolism , Ribonuclease, Pancreatic/toxicity , Ribonucleases/antagonists & inhibitors , Tumor Cells, Cultured , Tumor Stem Cell Assay
15.
Proteins ; 14(3): 392-400, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1438177

ABSTRACT

The P-30 protein (Onconase) of Rana pipiens oocytes and early embryos is homologous to members of the pancreatic ribonuclease superfamily and exhibits an antitumor activity in vitro and in vivo. It appears that the ribonucleolytic activity of P-30 protein may be required for its antitumor effects. A comparative molecular model of P-30 protein has been constructed based upon the known three-dimensional structure of bovine pancreatic RNase A in order to provide structural information. Functionally, these enzymes hydrolyze oligoribonucleotides to pyrimidine-3'-phosphate monoesters and 5'-OH ribonucleotides. In the modeling procedure, automated sequence alignments were revised based upon the inspection of the RNase A structure before the amino acids of the P-30 protein were assigned the coordinates of the RNase A template. The inevitable intermolecular steric clashes that result were relieved on an interactive graphics device through the adjustment of side chain torsion angles. This process was followed by energy minimization of the model, which served to optimize stereochemical geometry and to relieve any remaining unacceptably close contacts. The resulting model retains the essential features of RNase A as sequence insertions and deletions are almost exclusively found in exposed surface loops. The all atom superposition of active site residues of the P-30 protein model and an identically minimized RNase A structure has a root mean square deviation of 0.52 A. Though tentative, the model is consistent with a pyrimidine specificity.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Antineoplastic Agents/chemistry , Egg Proteins/chemistry , Rana pipiens/embryology , Ribonuclease, Pancreatic/chemistry , Ribonucleases/chemistry , Amino Acid Sequence , Animals , Cattle , Crystallization , Models, Molecular , Molecular Sequence Data , Sequence Alignment
16.
J Mol Biol ; 220(3): 739-55, 1991 Aug 05.
Article in English | MEDLINE | ID: mdl-1870129

ABSTRACT

Tetragonal and triclinic crystals of two ovomucoid inhibitor third domains from silver pheasant and Japanese quail, modified at their reactive site bonds Met18-Glu19 (OMSVP3*) and Lys18-Asp19 (OMJPQ3*), respectively, were obtained. Their molecular and crystal structures were solved using X-ray data to 2.5 A and 1.55 A by means of Patterson search methods using truncated models of the intact (virgin) inhibitors as search models. Both structures were crystallographically refined to R-values of 0.185 and 0.192, respectively, applying an energy restraint reciprocal space refinement procedure. Both modified inhibitors show large deviations from the intact derivatives only in the proteinase binding loops (Pro14 to Arg21) and in the amino-terminal segments (Leu1 to Val6). In the modified inhibitors the residues immediately adjacent to the cleavage site (in particular P2, P1, P1') are mobile and able to adapt to varying crystal environments. The charged end-groups, i.e. Met18 COO- and Glu19 NH3+ in OMSVP3*, and Lys18 COO- and Asp19 NH3+ in OMJPQ3*, do not form ion pairs with one another. The hydrogen bond connecting the side-chains of Thr17 and Glu19 (i.e. residues on either side of the scissile peptide bond) in OMSVP3 is broken in the modified form, and the hydrogen-bond interactions observed in the intact molecules between the Asn33 side-chain and the carbonyl groups of loop residues P2 and P1' are absent or weak in the modified inhibitors. The reactive site cleavage, however, has little effect on specific interactions within the protein scaffold such as the side-chain hydrogen bond between Asp27 and Tyr31 or the side-chain stacking of Tyr20 and Pro22. The conformational differences in the amino-terminal segment Leu1 to Val6 are explained by their ability to move freely, either to associate with segments of symmetry-related molecules under formation of a four-stranded beta-barrel (OMSVP3* and OMJPQ3) or to bind to surrounding molecules. Together with the results given in the accompanying paper, these findings probably explain why Khyd of small protein inhibitors of serine proteinases is generally found to be so small.


Subject(s)
Ovomucin/antagonists & inhibitors , Trypsin Inhibitor, Kazal Pancreatic/chemistry , Amino Acid Sequence , Animals , Binding Sites , Birds , Computer Simulation , Coturnix , Crystallization , Hydrolysis , Models, Molecular , Molecular Sequence Data , Protein Conformation , Trypsin Inhibitor, Kazal Pancreatic/isolation & purification , X-Ray Diffraction/methods
17.
J Mol Biol ; 220(4): 1041-53, 1991 Aug 20.
Article in English | MEDLINE | ID: mdl-1880798

ABSTRACT

We have measured equilibrium constants, Khyd, at pN 6 for the hydrolysis of the reactive site peptide bond (bond between residues 18 and 19) in 42 sequenced variants (39 natural, 3 semisynthetic) of avian ovomucoid third domains. The values range from 0.4 to approximately 35. In 35 cases the effect of a single amino acid replacement on Khyd could be calculated, 13 are without effect and 22 range from a factor of 1.25 to 5.5. Several, but not all, of the effects can be rationalized in terms of residue-residue interactions that are affected by the reactive site hydrolysis. As the measurements are very precise it appears that additional measurements on designed rather than natural variants should allow for the precise measurement of side-chain--side-chain interaction energies.


Subject(s)
Ovomucin/chemistry , Amino Acid Sequence , Animals , Birds , Glycosylation , Hydrogen-Ion Concentration , Hydrolysis , In Vitro Techniques , Kinetics , Molecular Sequence Data , Ovomucin/ultrastructure , Serine Endopeptidases/metabolism , Structure-Activity Relationship , Thermodynamics
18.
J Biol Chem ; 266(1): 245-51, 1991 Jan 05.
Article in English | MEDLINE | ID: mdl-1985896

ABSTRACT

Rana pipiens oocytes and early embryos contain large amounts of a basic protein with antiproliferative/cytotoxic activity against several tumor cell lines in vitro (Darzynkiewicz, Z., Carter, S. P., Mikulski, S. M., Ardelt, W., and Shogen, K. (1988) Cell Tissue Kinet. 21, 169-182; Mikulski, S.M., Viera, A., Ardelt, W., Menduke, H., and Shogen, K. (1990) Cell Tissue Kinet. 23, 237-246), as well as antitumor activity in vivo (Mikulski, S. M., Ardelt, W., Shogen, K., Bernstein, E. H., and Menduke, H. (1990) J. Natl. Cancer Inst. 82, 151-153). The protein, provisionally named P-30 Protein, was purified to homogeneity from early embryos and characterized. It is a single-chain protein consisting of 104 amino acid residues in the following sequence: less than Glu1-Asp-Trp-Leu-Thr-Phe-Gln-Lys-Lys-His-Ile-Thr-Asn-Thr- Arg15-Asp-Val-Asp-Cys-Asp-Ans-Ile-Met-Ser-Thr-Asn-Leu-Phe-His-C ys30-Lys-Asp-Lys - Asn-Thr-Phe-Ile-Tyr-Ser-Arg-Pro-Glu-Pro-Val-Lys45-Ala-Ile-Cys-Lys- Gly-Ile-Ile- Ala-Ser-Lys-Asn-Val-Leu-Thr-Thr60-Ser-Glu-Phe-Tyr-Leu-Ser-Asp -Cys-Asn-Val-Thr-Ser-Arg-Por-Cys75-Lys-Tyr-Lys-Leu-Lys-Lys-Ser-Thr -Asn-Lys-Phe- Cys-Val-Thr-Cys90-Glu-Asn-Gln-Ala-Pro-Val-His-Phe-Val-Gly-Val-Gly- Ser-Cys104-OH . Its molecular weight calculated from the sequence is 11,819. The sequence homology clearly indicates that the protein belongs to the superfamily of pancreatic ribonuclease. It is also demonstrated that it indeed exhibits a ribonucleolytic activity against highly polymerized RNA and that this activity seems to be essential for its antiproliferative/cytotoxic effects.


Subject(s)
Antineoplastic Agents/isolation & purification , Embryo, Nonmammalian/chemistry , Oocytes/chemistry , Proteins/isolation & purification , Agglutination , Amino Acid Sequence , Animals , Cell Line , Cell Survival/drug effects , Chromatography, Gel , Chromatography, Ion Exchange , Female , Humans , Molecular Sequence Data , Multigene Family , Pancreas/enzymology , Peptide Fragments/isolation & purification , Proteins/genetics , Proteins/pharmacology , Rana pipiens , Ribonucleases/genetics
19.
J Protein Chem ; 9(6): 715-25, 1990 Dec.
Article in English | MEDLINE | ID: mdl-2073323

ABSTRACT

Ovomucoids were isolated from 25 avian species other than the 101 studied in Laskowski et al. (1987, Biochemistry 26, 202-221). These were subjected to limited proteolysis with an appropriate enzyme, and connecting peptide extended ovomucoid third domains were isolated and sequenced to the end in a protein sequencer. Of the 25 new sequences, 13 duplicate ones were already known, and 12 are unique. Probably the most striking findings are a Pro14----Ser14 replacement in weka, an Ala14----Thr15 replacement in Bulwer's pheasant, the discovery of two additional amino acid residues Ile18 and Gly18 at the P1 reactive site position in Kalij pheasant and tawny frogmouth, respectively, and the first finding of a negative (Glu34) rather than positive (Lys34 or Arg34) amino acid residue at the NH2 terminus of the alpha helix in caracara ovomucoid third domain. These results complete the determination of all the sequences of ovomucoid third domains in the four species genus Gallus, in the five species genus Syrmaticus, and in the two species genera Aix and Pavo.


Subject(s)
Ovomucin/analysis , Peptide Fragments/analysis , Selection, Genetic , Amino Acid Sequence , Animals , Biological Evolution , Birds/genetics , Egg Proteins/genetics , Molecular Sequence Data , Ovomucin/genetics , Sequence Alignment
20.
Cell Tissue Kinet ; 23(3): 237-46, 1990 May.
Article in English | MEDLINE | ID: mdl-2357721

ABSTRACT

P-30 protein, a novel protein isolated in our laboratory from fertilized Rana pipiens eggs, has been shown to possess significant anti-proliferative and cytotoxic activity against a variety of human tumour cell lines. This protein also shows a potent anti-tumour activity in vivo in animal tumour models and is currently undergoing Phase I human clinical trials in cancer patient volunteers. The present study describes the in vitro effects of the concerted action of this protein and two other agents which affect the cell proliferative cycle. A significant potentiation of the P-30 protein-induced cell growth inhibition by tamoxifen as well as trifluoroperazine (Stelazine) in both the human A-549 lung carcinoma and the ASPC-1 pancreatic adenocarcinoma systems at wide ranges of drug concentrations was observed. The effect was apparently due to the synergistic action of P-30 protein and the agents tested. This data may provide clues that can be useful in explaining the mechanism of its anti-tumour activity. The results are also helpful for the designing in vivo animal and, perhaps eventually, human studies, whereby the combination therapies utilizing P-30 protein with agents of relatively low toxicity such as tamoxifen and/or Stelazine could offer a promising treatment(s) for these notoriously refractory types of human cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Lung Neoplasms/pathology , Pancreatic Neoplasms/pathology , Proteins/pharmacology , Ribonucleases , Tamoxifen/pharmacology , Trifluoperazine/pharmacology , Adenocarcinoma/pathology , Cell Division/drug effects , Drug Synergism , Humans , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...