Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Oncogene ; 31(31): 3584-96, 2012 Aug 02.
Article in English | MEDLINE | ID: mdl-22120720

ABSTRACT

Senescence is an irreversible growth arrest phenotype adopted by cells that has a key role in protecting organisms from cancer. There is now considerable interest in therapeutic strategies that reactivate this process to control the growth of cancer cells. Protein kinase-Cι (PKCι) is a member of the atypical PKC family and an important downstream mediator in the phosphoinositide-3-kinase (PI-3-kinase) pathway. PKCι expression was found to be upregulated in a subset of breast cancers and breast cancer cell lines. Activation of the PI-3-kinase pathway by introduction of mutant, oncogenic PIK3CA into breast mammary epithelial cells increased both the expression and activation of PKCι. In breast cancer cells lines overexpressing PKCι, depletion of PKCι increased the number of senescent cells, as assessed by senescence-associated ß-galactosidase, morphology and bromodeoxyuridine incorporation. This phenomenon was not restricted to breast cancer cells, as it was also seen in glioblastoma cells in which PKCι is activated by loss of PTEN. Senescence occurred in the absence of a detectable DNA-damage response, was dependent on p21 and was enhanced by the aurora kinase inhibitor VX-680, suggesting that senescence is triggered by defects in mitosis. Depletion of PKCι had no effect on senescence in normal mammary epithelial cell lines. We conclude that PKCι is overexpressed in a subset of cancers where it functions to suppress premature senescence. This function appears to be restricted to cancer cells and inhibition of PKCι may therefore be an effective way to selectively activate premature senescence in cancer cells.


Subject(s)
Breast Neoplasms/enzymology , Cellular Senescence/physiology , Isoenzymes/biosynthesis , Protein Kinase C/biosynthesis , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cellular Senescence/drug effects , Class I Phosphatidylinositol 3-Kinases , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , Glioblastoma/enzymology , Glioblastoma/pathology , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Piperazines/pharmacology , Transfection , Up-Regulation , beta-Galactosidase/analysis
2.
Oncogene ; 27(25): 3587-95, 2008 Jun 05.
Article in English | MEDLINE | ID: mdl-18212741

ABSTRACT

Glioblastoma multiforme is the most aggressive form of primary brain tumor and remains largely incurable, in large part, due to its highly invasive nature. The phosphoinositide (PI) 3-kinase pathway is often constitutively active in these tumors due to activating mutations in the epidermal growth factor receptor, or deletion/loss of function of the tumor suppressor PTEN. Protein kinase C type iota (PKC iota), a member of the atypical protein kinase C family, is activated by the PI 3-kinase pathway and is an important downstream mediator. Here, we have assessed the role of PKC iota in glioblastoma cell invasion. Depletion of PKC iota with RNA interference caused an increase in actin stress fibers and a decrease in cell motility and invasion. Gene expression microarray analysis of U87MG cells showed that PKC iota repressed expression of mRNA for RhoB, which has previously been shown to have a role in actin stress fiber formation. Western blot analysis showed that both PKC iota depletion and pharmacological inhibition of PKC iota caused an increase in the protein levels of RhoB, as did inhibition of PI 3-kinase. Expression of RhoB from a constitutive promoter caused changes in actin stress fibers and cell invasion that were similar to those seen with PKC iota depletion. These data show that PKC iota, activated as a consequence of aberrant upstream PI 3-kinase signaling, mediates glioblastoma cell motility and invasion, and that repression of RhoB is key downstream event in PKC iota signaling leading to enhanced cell motility. In addition, constitutive expression of RhoB repressed PKC iota activity, as assessed by its phosphorylation status on Thr555. PKC iota and RhoB are, therefore, mutually antagonistic, potentially creating a sensitive switch between invasive and non-invasive phenotypes.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Glioblastoma/pathology , Isoenzymes/metabolism , Protein Kinase C/metabolism , rhoB GTP-Binding Protein/metabolism , Cell Line, Tumor , Cell Movement , Gene Expression Profiling , Humans , Models, Biological , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic , RNA Interference
3.
Oncogene ; 25(20): 2909-19, 2006 May 11.
Article in English | MEDLINE | ID: mdl-16331246

ABSTRACT

Glioblastoma multiforme is an aggressive form of brain cancer that responds poorly to chemotherapy and is generally incurable. The basis for the poor response of this cancer to chemotherapy is not well understood. The atypical protein kinases C (PKCiota and PKCzeta) have previously been implicated in leukaemia cell chemoresistance. To assess the role of atypical PKC in glioblastoma cell chemoresistance, RNA interference was used to deplete human glioblastoma cells of PKCiota. Transfection of cells with either of two different RNA duplexes specific for PKCiota caused a partial sensitisation to cell death induced by the chemotherapy agent cisplatin. To screen for possible mechanisms for PKCiota-mediated chemoresistance, microarray analysis of gene expression was performed on RNA from glioblastoma cells that were either untreated or depleted of PKCiota. This identified sets of genes that were regulated either positively or negatively by PKCiota. Within the set of genes that were negatively regulated by PKCiota, the function of the gene coding for GMFbeta, an enhancer of p38 mitogen-activated protein kinase (MAP kinase) signaling, was investigated further, as the p38 MAP kinase pathway has been previously identified as a key mediator of cisplatin cytotoxicity. The expression of both GMFbeta mRNA and protein increased upon PKCiota depletion, and this was accompanied by an increase in cisplatin-activated p38 MAP kinase signaling. Transient overexpression of GMFbeta increased cisplatin-activated p38 MAP kinase signaling and also sensitised cells to cisplatin cytotoxicity. The increase in cisplatin cytotoxicity seen with PKCiota depletion was blocked by the p38 MAP kinase inhibitor SKF86002. These data show that PKCiota can confer partial resistance to cisplatin in glioblastoma cells by suppressing GMFbeta-mediated enhancement of p38 MAP kinase signaling.


Subject(s)
Antineoplastic Agents/toxicity , Cell Proliferation/drug effects , Cisplatin/toxicity , Glioblastoma/drug therapy , Isoenzymes/metabolism , Protein Kinase C/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Cytoprotection , Gene Expression Profiling , Glia Maturation Factor/metabolism , Glioblastoma/enzymology , Humans , Isoenzymes/genetics , Microarray Analysis , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Kinase C/genetics , Signal Transduction , Transfection , Tumor Cells, Cultured
4.
Invest New Drugs ; 23(2): 147-55, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15744591

ABSTRACT

PURPOSE: To define the maximum tolerated dose (MTD), the dose limiting toxicity (DLT), the biological active (BA) dose and the pharmacokinetics (PK) of the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor ZD1839 (Iressa) when administered continuously as a once daily dose in patients with advanced, incurable solid tumours. PATIENTS AND METHODS: Twenty-eight patients were enrolled in cohorts of three from three National Cancer Institute of Canada Clinical Trials Group (NCIC CTG) centers. ZD1839 was given at doses from 150 to 800 mg daily orally and patients underwent a pretreatment and a 28 day post treatment tumor biopsy, while PK sampling was performed on days 8, 15, 22, 29, and a toxicity assessment every 28 days. RESULTS: All twenty-eight patients were evaluable for non-hematological and hematological toxicity. Twenty-seven were evaluable for response. The MTD was not reached but DLT included reversible rash and diarrhea. One patient with urachal cancer had a transient 55% decrease in tumor size and two other patients (breast and non-small cell lung cancer) had minor responses; three additional patients had pharmacodynamic evidence of target effect. PK demonstrated steady state within the first 2 weeks of dosing and dose dependent exposure. CONCLUSION: It appears that ZD 1839 at a dose of 800 m/day was tolerable, although some patients required dose modification for diarrhea. Doses above 250 m/day demonstrate biologic activity and could be consider for future study in a variety of EGFR positive tumor types.


Subject(s)
Antineoplastic Agents/therapeutic use , ErbB Receptors/drug effects , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Administration, Oral , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Canada , ErbB Receptors/metabolism , Female , Gefitinib , Half-Life , Humans , Male , Middle Aged , Neoplasms/pathology , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Quinazolines/adverse effects , Quinazolines/pharmacokinetics
5.
Curr Cancer Drug Targets ; 2(2): 91-102, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12188912

ABSTRACT

The epidermal growth factor (EGF) receptor is overexpressed in many cancers, and is under intensive investigation as a target for cancer therapy. Cancer cells have also been shown to express mutated EGF receptors; these are potentially highly specific targets for cancer therapeutics, as they have not been detected in any normal adult tissues. The most common of these mutant EGF receptors, EGFRvIII, is one in which amino acids 6 - 273 of the extracellular domain are deleted. This specific mutation is common in glioblastoma and in several other types of cancer, and has been shown to promote aggressive growth of tumors in vivo. The loss of part of the extracellular domain results in a receptor that has constitutive tyrosine kinase activity. Current evidence suggests that EGFRvIII has altered signalling properties compared to normal EGF receptor. The mutation in EGFRvIII also creates a new, cancer cell-specific epitope. This epitope is extracellular and therefore represents a very promising target for antibody-directed therapeutics. This review covers our current understanding of the properties of EGFRvIII, and recent developments in the characterization and therapeutic application of EGFRvIII-specific antibodies.


Subject(s)
Antineoplastic Agents/pharmacology , ErbB Receptors/drug effects , ErbB Receptors/genetics , Neoplasms/drug therapy , Amino Acid Sequence , Animals , ErbB Receptors/immunology , Gene Targeting , Humans , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Radiopharmaceuticals , Signal Transduction/drug effects
6.
Biochim Biophys Acta ; 1538(1): 1-9, 2001 Feb 05.
Article in English | MEDLINE | ID: mdl-11341977

ABSTRACT

Glioblastoma cells express a mutant EGF receptor (EGFRvIII) that has constitutive tyrosine kinase activity and enhances their tumorigenicity. Here we show that EGFRvIII promotes constitutive phosphorylation of extracellular regulated kinases (ERKs) in glioblastoma cells in the absence of EGF. EGFRvIII also promoted constitutive activation of phosphoinositide 3-kinase in these cells, as assessed by phosphorylation of protein kinase B/akt. As expected, phosphorylation of protein kinase B/akt was blocked by the phosphoinositide 3-kinase inhibitors wortmannin and LY294002. Less expectedly, we found that this treatment also blocked EGFRvIII-induced phosphorylation of ERKs. In contrast, ERK phosphorylation induced by EGF-activated normal EGF receptor in the same cells was largely unaffected by treatment with phosphoinositide 3-kinase inhibitors. This difference in behavior between the normal receptor and EGFRvIII was not due to differences in the levels of activated EGFRvIII and wild-type EGF receptor, as the two types of receptor were tyrosine phosphorylated to a similar extent under the experimental conditions used. EGFRvIII activation of ERKs was also sensitive to the phospholipase C inhibitor U73122, whereas ERK activation by normal EGF receptor was not. These results show that EGFRvIII and wild-type EGF receptor preferentially use different signaling pathways to induce ERK phosphorylation. The different mechanisms of ERK activation used by normal and mutant EGF receptors may be important in understanding the potent tumorigenic activity of EGFRvIII.


Subject(s)
ErbB Receptors/metabolism , Mitogen-Activated Protein Kinases/metabolism , Dimethyl Sulfoxide , Enzyme Activation , Enzyme Inhibitors/pharmacology , ErbB Receptors/genetics , Mitogen-Activated Protein Kinases/analysis , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mutation , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Quinazolines , Tumor Cells, Cultured , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism , Tyrphostins/pharmacology
7.
J Mol Biol ; 308(5): 883-93, 2001 May 18.
Article in English | MEDLINE | ID: mdl-11352579

ABSTRACT

Epitope mapping studies and the determination of the structure to 1.8 A resolution have been carried out for the antigen-binding fragment MR1 in complex with peptide antigen. MR1 is specific for the novel fusion junction of the mutant epidermal growth factor receptor EGFRvIII and has been reported to have a high degree of specificity for the mutant EGFRvIII over the wild-type EGF receptor. The structure of the complex shows that the peptide antigen residue side-chains found by epitope mapping studies to be critical for recognition are accommodated in pockets on the surface of the Fv. However, the most distinctive portion of the peptide antigen, the novel fusion glycine residue, makes no contact to the Fv and does not contribute directly to the epitope. The specificity of MR1 lies in the ability of this glycine residue to assume the restricted conformation needed to form a type II' beta-hairpin turn more easily, and demonstrates that a peptide antigen can be used to generate a conformational epitope.


Subject(s)
ADP Ribose Transferases , Antibody Specificity , Bacterial Toxins , Epitope Mapping , Epitopes/chemistry , ErbB Receptors/chemistry , ErbB Receptors/immunology , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/immunology , Virulence Factors , Amino Acid Sequence , Antigens/chemistry , Antigens/immunology , Base Sequence , Crystallography, X-Ray , Disulfides/metabolism , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , ErbB Receptors/genetics , Exotoxins/chemistry , Exotoxins/genetics , Exotoxins/metabolism , Hydrogen Bonding , Immunoglobulin Fragments/genetics , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Peptide Fragments/immunology , Protein Conformation , Protein Engineering , Pseudomonas , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Temperature , Thermodynamics , Pseudomonas aeruginosa Exotoxin A
8.
J Immunol Methods ; 237(1-2): 147-57, 2000 Apr 03.
Article in English | MEDLINE | ID: mdl-10725459

ABSTRACT

We have investigated targeting of retroviral vectors to a mutant EGF receptor (EGFRvIII) that is expressed in cancers of the brain, breast, lung and ovary, but is not found in any normal tissues. An expression plasmid was made in which a single chain Fv antibody specific for EGFRvIII was inserted at a novel position within a disulphide-bonded surface loop near the native receptor binding site of the Moloney leukemia virus ecotropic envelope glycoprotein. This fusion protein was expressed and incorporated into retroviral particles as efficiently as normal envelope glycoprotein. Retroviral vectors made with the fusion protein were able to bind peptide antigen and EGFRvIII expressed on the surface of human glioblastoma cells. The retroviral vectors had normal levels of infectivity on mouse cells, showing that the envelope glycoprotein tolerated a large insertion at this site, but did not show significant infectivity to human cells expressing EGFRvIII. Thus we were able to redirect retrovirus binding to this tumour-specific target without perturbing the normal function of the ecotropic envelope glycoprotein, but this was not sufficient to mediate infectivity via this receptor.


Subject(s)
ErbB Receptors/genetics , Genetic Vectors , Immunoglobulin Variable Region/genetics , Neoplasms/therapy , Retroviridae/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Binding Sites/genetics , DNA Primers/genetics , ErbB Receptors/metabolism , Female , Genetic Therapy , Humans , Immunoglobulin Variable Region/chemistry , Leukemia Virus, Murine/genetics , Mice , Models, Molecular , Molecular Sequence Data , Neoplasms/genetics , Neoplasms/metabolism , Protein Conformation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
9.
Clin Cancer Res ; 5(9): 2646-52, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10499644

ABSTRACT

The incidence of neoplastic meningitis is on the rise. Neoplastic meningitis can result from a direct seeding of the neuraxis by primary brain tumors or by hematogeneous spread of systemic solid tumors. A frequent genetic alteration in primary brain tumors such as gliomas is an in-frame deletion in the epidermal growth factor receptor (EGFR) gene EGFRvIII, which brings together what were normally distant polypeptide sequences in the intact receptor. A novel glycine is formed at the fusion junction, resulting in a unique and tumor-specific target. By using phage display, we have isolated a single-chain antibody specific for the EGFRvIII mutation and expressed it with a modified form of the Pseudomonas exotoxin to form the immunotoxin MR1scFvPE38KDEL (MR-1). The multiple dose toxicity and therapeutic efficacy of MR-1 immunotoxin were tested in an athymic rat model of neoplastic meningitis. The maximally tolerated doses in non-tumor-bearing rats were three doses of 3 microg each. For therapeutic studies, the target was a neoplastic meningitis induced by intrathecal inoculation of the EGFRvIII-expressing human glioma U87MG.deltaEGFR. A dose escalation study compared the survival of three equal doses of 1, 2, and 3 microg of MR-1 immunotoxin with saline or 3 microg of the control immunotoxin specific for the interleukin 2 receptor, anti-Tac. All animals treated with three doses of saline or 3 microg of anti-Tac died, with median survival of 7 and 10 days, respectively. There were 75% (six of eight) long-term survivors in the group treated with three doses of 1 microg and 57% (four of seven) long-term survivors in the groups treated with three doses of either 2 or 3 microg of MR-1 immunotoxin. None of the MR-1 immunotoxin-treated groups reached median survival by the termination of the study at 53 days. Therefore, median survival was estimated to be >53 days, resulting in an estimated increase in median survival of >657% compared with saline and 430% versus anti-Tac. Compartmental therapy with three doses of 2 microg of MR-1 immunotoxin is effective in the treatment of EGFRvIII-expressing neoplastic meningitis. This dose was found to have no clinical or histopathological effects on non-tumor-bearing animals. MR-1 immunotoxin is, therefore, considered specific and safe within its therapeutic window. Phase I clinical trials for tumors invading the intrathecal space that express the EGFRvIII target should be initiated.


Subject(s)
ADP Ribose Transferases , Bacterial Toxins , ErbB Receptors/immunology , ErbB Receptors/metabolism , Exotoxins/pharmacology , Immunoglobulin Variable Region/pharmacology , Immunotoxins/pharmacology , Meningeal Neoplasms/drug therapy , Virulence Factors , Animals , Antibody Specificity , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Exotoxins/toxicity , Female , Humans , Immunoglobulin Variable Region/toxicity , Immunotoxins/toxicity , Injections, Spinal , Meningeal Neoplasms/metabolism , Mice , Mice, Nude , Mutation , Protein Synthesis Inhibitors/pharmacology , Rats , Rats, Nude , Tumor Cells, Cultured , Pseudomonas aeruginosa Exotoxin A
10.
Clin Cancer Res ; 5(6): 1539-49, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10389943

ABSTRACT

A single-chain antibody fragment, MR1(scFv), with specific binding to epidermal growth factor receptor-vIII (EGFRvIII), was produced, radiolabeled, and evaluated for biodistribution in human glioma-bearing athymic mice. The mutant receptor EGFRvIII has a deletion in its extracellular domain that results in the formation of a new, tumor-specific antigen found in glioblastomas, breast carcinomas, and other tumors. The scFv molecule, designed as V(H)-(Gly4-Ser)3-V(L), was expressed in Escherichia coli in inclusion body form; recovered scFv fragments were properly refolded in redox-shuffling buffer. Size-exclusion chromatography of purified scFv demonstrated a protein monomer of Mr 26,000. Labeling was performed using N-succinimidyl 5-[125I]iodo-3-pyridinecarboxylate (SIPC) or Iodogen to specific activities of 0.5-2.0 mCi/mg, with yields of 35-50% and 45-70%, respectively. The immunoreactive fraction (IRF) of the labeled MR1(scFv) was 65-80% when SIPC was used and 50-55% when Iodogen was used. The affinity (K(A)) of MRI(scFv) for EGFRvIII was 4.3 x 10(7) +/- 0.1 x 10(7) M(-1) by BIAcore analysis, and it was 1.0 x 10(8) +/- 0.1 x 10(8) M(-1) and by Scatchard analysis versus EGFRvIII-expressing cells. After incubation at 37 degrees C for 24 h, the binding affinity was maintained, and the IRF was maintained at 60-70%. The specificity of MR1(scFv) for EGFRvIII was demonstrated in vitro by incubation of radiolabeled MR1(scFv) with the EGFRvIII-expressing U87MG.deltaEGFR cell line in the presence or absence of competing unlabeled MR1(scFv) or anti-EGFRvIII MAbs L8A4 and H10. In biodistribution studies using athymic mice bearing s.c. U87MG.deltaEGFR tumor xenografts, animals received intratumoral or i.v. infusions of paired-label [125I]SIPC-MR1(scFv) and [131I]SIPC-anti-Tac(scFv) as a control. When given by the intratumoral route, MR1(scFv) retained high tumor uptakes of 85% injected dose per gram of tissue at 1 h and 16% injected dose per gram of tissue at 24 h following administration. Specific: control scFv tumor uptake ratios of more than 20:1 at 24 h demonstrated specific localization of MR1(scFv). The excellent tumor retention of MR1(scFv), combined with its rapid clearance from normal tissues, resulted in high tumor:normal organ ratios.


Subject(s)
Antibody Specificity , ErbB Receptors/immunology , Glioma/therapy , Immunoglobulin Variable Region/pharmacology , Amino Acid Sequence , Animals , Binding, Competitive , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , ErbB Receptors/metabolism , Female , Glioma/immunology , Glioma/metabolism , Humans , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/metabolism , Iodine Radioisotopes , Mice , Molecular Sequence Data , Neoplasm Transplantation , Protein Folding , Tissue Distribution
11.
Proc Natl Acad Sci U S A ; 93(25): 14815-20, 1996 Dec 10.
Article in English | MEDLINE | ID: mdl-8962138

ABSTRACT

EGFRvIII is a mutant epidermal growth factor receptor found in glioblastoma, and in carcinoma of the breast, ovary, and lung. The mutant receptor has a deletion in its extracellular domain that results in the formation of a new, tumor-specific extracellular sequence. Mice were immunized with a synthetic peptide corresponding to this sequence and purified EGFRvIII. A single chain antibody variable domain (scFv) phage display library of 8 x 10(6) members was made from the spleen of one immunized mouse. A scFv specific for EGFRvIII was isolated from this library by panning with successively decreasing amounts of synthetic peptide. This was used to make an immunotoxin by fusing the scFv DNA sequence to sequences coding for domains II and III of Pseudomonas exotoxin A. Purified immunotoxin had a Kd of 22 nM for peptide and a Kd of 11 nM for cell-surface EGFRvIII. The immunotoxin was very cytotoxic to cells expressing EGFRvIII, with an IC50 of 1 ng/ml (16 pM) on mouse fibroblasts transfected with EGFRvIII and an IC50 of 7-10 ng/ml (110-160 pM) on transfected glioblastoma cells. There was no cytotoxic activity at 1000 ng/ml on the untransfected parent glioblastoma cell line. The immunotoxin was completely stable upon incubation at 37 degrees C for 24 h in human serum. The combination of good affinity, cytotoxicity and stability make this immunotoxin a candidate for further preclinical evaluation.


Subject(s)
ErbB Receptors/immunology , Glioblastoma/immunology , Immunoglobulin Variable Region/immunology , Immunotoxins/immunology , Animals , ErbB Receptors/genetics , Gene Library , Glioblastoma/metabolism , Humans , Immunoglobulin Variable Region/pharmacology , Immunotoxins/genetics , Immunotoxins/pharmacology , Mice , Molecular Sequence Data , Mutation , Tumor Cells, Cultured
12.
Clin Cancer Res ; 1(8): 859-64, 1995 Aug.
Article in English | MEDLINE | ID: mdl-9816055

ABSTRACT

Human cancers arise from a series of mutations, many of which direct the expression of mutant proteins with altered functions. These aberrant proteins are attractive targets for new therapeutic agents. One such protein is a mutant epidermal growth factor receptor (EGFRvIII) that has an in-frame deletion near the NH2 terminus of its extracellular domain. This protein was first identified in human gliomas, but has also been shown to be present in lung and breast carcinomas. The deletion results in a receptor with constitutive tyrosine kinase activity that enhances the tumorigenicity of glioblastomas in vivo. The deletion also creates a tumor-specific cell-surface sequence at the deletion junction. Three specific anti-EGFRvIII mAbs have been isolated following immunization with a mixture of a deletion junction synthetic peptide and EGFRvIII as present on cell membranes. We have constructed immunotoxins by conjugating a modified version of Pseudomonas exotoxin A to these mAbs. Immunotoxins were tested on cells that had been transfected with cDNA for the EGFRvIII receptor and expressed receptor protein at 5 x 10(5) receptors/cell. All three immunotoxins were cytotoxic to these cells, with 50% inhibition of protein synthesis occurring in a 15-50 pM range. The immunotoxins specifically targeted EGFRvIII, as their cytotoxicity could be blocked by their respective free antibody. They showed little or no cytotoxicity to cells expressing high levels of normal epidermal growth factor receptors, demonstrating that they are able to discriminate between cells expressing the mutant receptor and those expressing the wild-type receptor. Immunotoxins targeted to mutant epidermal growth factor receptors are promising candidates for further development as tumor cell-specific therapeutic agents.


Subject(s)
Cell Survival/drug effects , ErbB Receptors/drug effects , ErbB Receptors/genetics , Immunotoxins/toxicity , 3T3 Cells , Animals , Antibodies, Monoclonal , Breast Neoplasms/genetics , Drug Design , Female , Glioma/genetics , Humans , Immunotoxins/chemistry , Lung Neoplasms/genetics , Mice , Mutagenesis, Site-Directed , Oligopeptides , Protein Sorting Signals , Recombinant Proteins/biosynthesis , Recombinant Proteins/drug effects , Sequence Deletion , Transfection
14.
J Biol Chem ; 269(12): 8680-5, 1994 Mar 25.
Article in English | MEDLINE | ID: mdl-7510696

ABSTRACT

We have been studying cAMP signaling in L6 myoblasts because of its potential role in regulating the differentiation of these cells into multinucleate myotubes. Previous studies have shown that treatment of L6 myoblasts with cAMP analogs causes an increase in cAMP phosphodiesterase activity. To assess the role of protein kinase A in this cAMP-mediated increase in cAMP phosphodiesterase activity, L6 myoblasts were transfected with a plasmid containing the cDNA for a mutant regulatory subunit of protein kinase A, which functions as a dominant negative inhibitor of this enzyme. The cDNA was under control of the metallothionein promoter in the construct. Induction of the mutant regulatory subunit with Zn2+ decreased cAMP-dependent protein kinase activity by 90%. Zn2+ treatment was also able to completely block the cAMP-mediated increase in phosphodiesterase activity, showing that this effect is mediated by protein kinase A. The activity of the cAMP-induced phosphodiesterase was inhibited by low concentrations of RO 20-1724, showing that it was a member of the type IV low Km cAMP phosphodiesterase family of enzymes. We used the polymerase chain reaction and consensus primers designed to amplify phosphodiesterase sequences to show that L6 myoblasts also contain mRNA for a type IV low Km cAMP phosphodiesterase designated PDE3.1. The levels of this mRNA were increased greatly by treatment with dibutyryl cAMP or forskolin in L6 myoblasts and also in differentiated L6 myotubes. Run-off transcription assays showed that this increase in PDE mRNA was regulated, at least in part, by an increase in the rate of transcription of the PDE3 gene. The induction of PDE3 message by cAMP was blocked when the L6 transfectants were treated with Zn2+ to induce protein kinase A inhibition. Therefore, some of the cAMP-mediated increase in phosphodiesterase activity seen in L6 myoblasts is due to a protein kinase A-mediated increase in PDE3 mRNA. This pathway may serve as a feedback mechanism to modulate the inhibitory effects of cAMP on myogenesis.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Muscles/enzymology , 1-Methyl-3-isobutylxanthine/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Animals , Base Sequence , Bucladesine/pharmacology , Cell Differentiation , Cell Line , Colforsin/pharmacology , DNA Primers/chemistry , Gene Expression , Molecular Sequence Data , Muscles/cytology , RNA, Messenger/genetics , Rats
15.
J Cell Biol ; 121(1): 193-9, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8458868

ABSTRACT

A potential regulatory linkage between the biosynthesis of colligin, a collagen-binding protein of the ER, and procollagen I was examined under a variety of experimental conditions. Cell lines which did not produce a significant amount of procollagen I mRNA also lacked the capacity to produce colligin mRNA. Anchorage-dependent cell lines like L6 myoblasts and normal rat kidney fibroblasts produced both colligin and procollagen I mRNA, but the level of both was concurrently reduced considerably in their ras-transformed counterparts. Similarly, during the differentiation of L6 myoblasts, levels of both colligin and procollagen declined together. Treatment of myoblasts by dexamethasone or EGF led to a decrease in the steady-state levels of procollagen I mRNA, and this was, again, accompanied by a decrease in colligin mRNA synthesis. On the other hand, when the rate of procollagen I synthesis was stimulated by treatment of myoblasts with TGF beta, it led to the concurrent augmentation of both the mRNA and protein levels of colligin. A linkage between the regulation of synthesis of procollagen I and colligin thus seems to exist. The only exception to this generalization is provided by the heat induction behavior of the two proteins. Treatment of myoblasts for a very short period leads to an increase in the synthesis of both the mRNA and protein levels of colligin. This, however, is not accompanied by a change in the mRNA levels of procollagen I. These studies establish that colligin and procollagen are generally tightly co-regulated except after heat shock, suggesting an important functional linkage.


Subject(s)
Carrier Proteins/metabolism , Collagen/metabolism , Procollagen/metabolism , Serine Proteinase Inhibitors/metabolism , Animals , Cell Differentiation , Cells, Cultured , Collagen/biosynthesis , Dexamethasone/pharmacology , Epidermal Growth Factor/physiology , Glycoproteins , Heat-Shock Proteins/metabolism , Humans , Mice , Muscles/cytology , Muscles/metabolism , RNA, Messenger/genetics , Rats , Transforming Growth Factor beta/physiology
16.
Biotechniques ; 12(4): 536-43, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1503756

ABSTRACT

A simple and rapid screening procedure was developed to study the interaction of the S. cerevisiae alpha 2 repressor with its operator sequence. An E. coli expression vector was constructed in which the alpha 2 coding sequence was placed under control of the lac promoter. Bacterial colonies containing this vector could be lysed and assayed directly for binding of wild-type and mutant operator sequences when grown on nitrocellulose filters. alpha 2 assayed in this way showed the same sequence specificity as determined in vivo. Pools of mutant alpha 2 repressors in which the codons for Arg185 or Ser181 in the homeodomain region were randomized were created by cassette mutagenesis. These pools of mutants were screened with the wild-type operator sequence to determine allowed amino acid substitutions at each position. Results suggest that both Arg185 and Ser181 have a role in high affinity operator binding.


Subject(s)
Escherichia coli/genetics , Gene Expression Regulation, Fungal , Genes, Regulator/physiology , Operator Regions, Genetic/physiology , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , Base Sequence , Genes, Bacterial/genetics , Genes, Fungal/genetics , Genetic Vectors , Molecular Sequence Data , Mutagenesis, Insertional , Mutagenesis, Site-Directed , Oligonucleotides , Plasmids
17.
Biochem J ; 264(1): 305-8, 1989 Nov 15.
Article in English | MEDLINE | ID: mdl-2604715

ABSTRACT

We showed previously that the levels of type I regulatory subunit of cyclic AMP-dependent protein kinase increase during differentiation of L6 skeletal myoblasts as a result of a specific decrease in its rate of degradation. Studies on the rates of degradation of the catalytic subunit show that unlike the type I regulatory subunit, catalytic subunit is degraded very slowly in myoblasts (t1/2 = 29 h) and more rapidly in myotubes (t1/2 = 14 h). As with the regulatory subunit, the degradation of catalytic subunit is increased by treatment of myoblasts with cyclic AMP analogues. These results suggest that the overall increase in the amount of type I cyclic AMP-dependent protein kinase holoenzyme during myogenesis is due to the increase in levels of mRNA for the catalytic subunit. This probably leads to an increase in the amount of catalytic subunit, which then stabilizes the regulatory subunit, thereby causing an increase in the levels of this protein also.


Subject(s)
Muscles/enzymology , Protein Kinases/metabolism , Animals , Blotting, Northern , Cell Differentiation , Cell Line , Gene Expression Regulation, Enzymologic , Macromolecular Substances , Muscles/cytology , Protein Kinases/genetics , RNA, Messenger/metabolism , Rats
18.
J Biol Chem ; 262(35): 17200-5, 1987 Dec 15.
Article in English | MEDLINE | ID: mdl-2445756

ABSTRACT

The regulation of type I cAMP-dependent protein kinase during the differentiation of L6 myoblasts has been investigated in order to assess a possible role for this enzyme in the control of myogenesis. Immunoblot analysis showed that the levels of the type I cAMP-dependent protein kinase regulatory subunit (RI) increased 3-fold during differentiation. However, measurement of RI mRNA levels using an RI cDNA probe showed that this increase was not regulated transcriptionally. Determinations of the turnover rate of RI demonstrated that the subunit becomes 3-fold more stable in the differentiated cells. Therefore, it appears that the increase is due to a decrease in the rate of degradation of RI. The increase in both the amount and stability of RI could be reversed by treating myotubes with cAMP analogues or forskolin. Furthermore, during differentiation there was a large decrease in cAMP levels. It was, therefore, concluded that the increase in RI levels seen during differentiation is probably due to an increase in the stability of the subunit as a result of a decrease in intracellular cAMP concentrations.


Subject(s)
Muscle Development , Protein Kinases/metabolism , Animals , Cell Differentiation , Cells, Cultured , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , DNA/analysis , Half-Life , Macromolecular Substances , Muscles/enzymology , RNA/analysis , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...