113 results on '"Jackson B. Gibbs"'
Search Results
2. Supplementary Figure 4 from MK-2461, a Novel Multitargeted Kinase Inhibitor, Preferentially Inhibits the Activated c-Met Receptor
- Author
-
Christopher J. Dinsmore, Nancy E. Kohl, Qinwen Zeng, Alexander A. Szewczak, John F. Reilly, Xianlu Qu, Cloud P. Paweletz, Bart A. Lutterbach, Wei Lu, Kaiko Kunii, Jason D. Katz, Ilona Kariv, James P. Jewell, Harold Hatch, Gaozhen Hang, Susana Gil, Jackson B. Gibbs, Sujal V. Deshmukh, Lenora J. Davis, An Chi, Melissa Chenard, Grace K.Y. Chan, and Bo-Sheng Pan
- Abstract
Supplementary Figure 4 from MK-2461, a Novel Multitargeted Kinase Inhibitor, Preferentially Inhibits the Activated c-Met Receptor
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Figure 2 from MK-2461, a Novel Multitargeted Kinase Inhibitor, Preferentially Inhibits the Activated c-Met Receptor
- Author
-
Christopher J. Dinsmore, Nancy E. Kohl, Qinwen Zeng, Alexander A. Szewczak, John F. Reilly, Xianlu Qu, Cloud P. Paweletz, Bart A. Lutterbach, Wei Lu, Kaiko Kunii, Jason D. Katz, Ilona Kariv, James P. Jewell, Harold Hatch, Gaozhen Hang, Susana Gil, Jackson B. Gibbs, Sujal V. Deshmukh, Lenora J. Davis, An Chi, Melissa Chenard, Grace K.Y. Chan, and Bo-Sheng Pan
- Abstract
Supplementary Figure 2 from MK-2461, a Novel Multitargeted Kinase Inhibitor, Preferentially Inhibits the Activated c-Met Receptor
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Figure 1 from MK-2461, a Novel Multitargeted Kinase Inhibitor, Preferentially Inhibits the Activated c-Met Receptor
- Author
-
Christopher J. Dinsmore, Nancy E. Kohl, Qinwen Zeng, Alexander A. Szewczak, John F. Reilly, Xianlu Qu, Cloud P. Paweletz, Bart A. Lutterbach, Wei Lu, Kaiko Kunii, Jason D. Katz, Ilona Kariv, James P. Jewell, Harold Hatch, Gaozhen Hang, Susana Gil, Jackson B. Gibbs, Sujal V. Deshmukh, Lenora J. Davis, An Chi, Melissa Chenard, Grace K.Y. Chan, and Bo-Sheng Pan
- Abstract
Supplementary Figure 1 from MK-2461, a Novel Multitargeted Kinase Inhibitor, Preferentially Inhibits the Activated c-Met Receptor
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Figure 5 from MK-2461, a Novel Multitargeted Kinase Inhibitor, Preferentially Inhibits the Activated c-Met Receptor
- Author
-
Christopher J. Dinsmore, Nancy E. Kohl, Qinwen Zeng, Alexander A. Szewczak, John F. Reilly, Xianlu Qu, Cloud P. Paweletz, Bart A. Lutterbach, Wei Lu, Kaiko Kunii, Jason D. Katz, Ilona Kariv, James P. Jewell, Harold Hatch, Gaozhen Hang, Susana Gil, Jackson B. Gibbs, Sujal V. Deshmukh, Lenora J. Davis, An Chi, Melissa Chenard, Grace K.Y. Chan, and Bo-Sheng Pan
- Abstract
Supplementary Figure 5 from MK-2461, a Novel Multitargeted Kinase Inhibitor, Preferentially Inhibits the Activated c-Met Receptor
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figure 1 Legend from Lung Cancer Cell Lines Harboring MET Gene Amplification Are Dependent on Met for Growth and Survival
- Author
-
Bo-Sheng Pan, Jackson B. Gibbs, Nancy E. Kohl, Gaozhen Hang, Harold Hatch, Lenora J. Davis, Qinwen Zeng, and Bart Lutterbach
- Abstract
Supplementary Figure 1 Legend from Lung Cancer Cell Lines Harboring MET Gene Amplification Are Dependent on Met for Growth and Survival
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Methods from Lung Cancer Cell Lines Harboring MET Gene Amplification Are Dependent on Met for Growth and Survival
- Author
-
Bo-Sheng Pan, Jackson B. Gibbs, Nancy E. Kohl, Gaozhen Hang, Harold Hatch, Lenora J. Davis, Qinwen Zeng, and Bart Lutterbach
- Abstract
Supplementary Methods from Lung Cancer Cell Lines Harboring MET Gene Amplification Are Dependent on Met for Growth and Survival
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Information from Lung Cancer Cell Lines Harboring MET Gene Amplification Are Dependent on Met for Growth and Survival
- Author
-
Bo-Sheng Pan, Jackson B. Gibbs, Nancy E. Kohl, Gaozhen Hang, Harold Hatch, Lenora J. Davis, Qinwen Zeng, and Bart Lutterbach
- Abstract
Supplementary Information from Lung Cancer Cell Lines Harboring MET Gene Amplification Are Dependent on Met for Growth and Survival
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Methods, Table 1, Figure Legends 1-5 from MK-2461, a Novel Multitargeted Kinase Inhibitor, Preferentially Inhibits the Activated c-Met Receptor
- Author
-
Christopher J. Dinsmore, Nancy E. Kohl, Qinwen Zeng, Alexander A. Szewczak, John F. Reilly, Xianlu Qu, Cloud P. Paweletz, Bart A. Lutterbach, Wei Lu, Kaiko Kunii, Jason D. Katz, Ilona Kariv, James P. Jewell, Harold Hatch, Gaozhen Hang, Susana Gil, Jackson B. Gibbs, Sujal V. Deshmukh, Lenora J. Davis, An Chi, Melissa Chenard, Grace K.Y. Chan, and Bo-Sheng Pan
- Abstract
Supplementary Methods, Table 1, Figure Legends 1-5 from MK-2461, a Novel Multitargeted Kinase Inhibitor, Preferentially Inhibits the Activated c-Met Receptor
- Published
- 2023
- Full Text
- View/download PDF
10. Data from Lung Cancer Cell Lines Harboring MET Gene Amplification Are Dependent on Met for Growth and Survival
- Author
-
Bo-Sheng Pan, Jackson B. Gibbs, Nancy E. Kohl, Gaozhen Hang, Harold Hatch, Lenora J. Davis, Qinwen Zeng, and Bart Lutterbach
- Abstract
Recent clinical successes of small-molecule epidermal growth factor receptor (EGFR) inhibitors in treating advanced non–small cell lung cancer (NSCLC) have raised hopes that the identification of other deregulated growth factor pathways in NSCLC will lead to new therapeutic options for NSCLC. Met, the receptor for hepatocyte growth factor, has been implicated in growth, invasion, and metastasis of many tumors including NSCLC. To assess the functional role for Met in NSCLC, we evaluated a panel of nine lung cancer cell lines for Met gene amplification, Met expression, Met pathway activation, and the sensitivity of the cell lines to short hairpin RNA (shRNA)–mediated Met knockdown. Two cell lines, EBC-1 and H1993, showed significant Met gene amplification and overexpressed Met receptors which were constitutively phosphorylated. The other seven lines did not exhibit Met amplification and expressed much lower levels of Met, which was phosphorylated only on addition of hepatocyte growth factor. We also found a strong up-regulation of tyrosine phosphorylation in β-catenin and p120/δ-catenin in the Met-amplified EBC-1 and H1993 cell lines. ShRNA-mediated Met knockdown induced significant growth inhibition, G1-S arrest, and apoptosis in EBC-1 and H1993 cells, whereas it had little or no effect on the cell lines that do not have Met amplification. These results strongly suggest that Met amplification identifies a subset of NSCLC likely to respond to new molecular therapies targeting Met. [Cancer Res 2007;67(5):2081–8]
- Published
- 2023
- Full Text
- View/download PDF
11. Proceedings from the 2009 genetic syndromes of the Ras/MAPK pathway: From bedside to bench and back
- Author
-
Jackson B. Gibbs, Katherine A. Rauen, Allan Balmain, Amy E. Roberts, Alcino J. Silva, Angela E. Lin, Benjamin D. Yu, Benjamin G. Neel, Richard R. Drake, Bruce R. Korf, Suzanne Schubbert, E. Elizabeth Patton, John C. Carey, Garry P. Nolan, Kevin Shannon, Carmen Guerra, James A. Fagin, Giovanni Neri, Bruce D. Gelb, Martin McMahon, Karen W. Gripp, Mark W. Kieran, Teri Melese, Randi J Hagerman, David A. Stevenson, David Viskochil, Lisa Schoyer, Marco Tartaglia, Judith Allanson, Eric Legius, Judith S. Sebolt-Leopold, Roger J. Packer, David H. Gutmann, Frank McCormick, Martin Zenker, Gideon Bollag, and Yoko Aoki
- Subjects
Genetics ,Legius syndrome ,MAPK/ERK pathway ,Mechanism (biology) ,business.industry ,RASopathy ,medicine.disease ,Bioinformatics ,LEOPARD Syndrome ,Article ,humanities ,Costello syndrome ,medicine ,Noonan syndrome ,Neurofibromatosis ,business ,health care economics and organizations ,Genetics (clinical) - Abstract
The RASopathies are a group of genetic syndromes caused by germline mutations in genes that encode components of the Ras/mitogen-activated protein kinase (MAPK) pathway. Some of these syndromes are neurofibromatosis type 1, Noonan syndrome, Costello syndrome, cardio-facio-cutaneous syndrome, LEOPARD syndrome and Legius syndrome. Their common underlying pathogenetic mechanism brings about significant overlap in phenotypic features and includes craniofacial dysmorphology, cardiac, cutaneous, musculoskeletal, GI and ocular abnormalities, and a predisposition to cancer. The proceedings from the symposium “Genetic Syndromes of the Ras/MAPK Pathway: From Bedside to Bench and Back” chronicle the timely and typical research symposium which brought together clinicians, basic scientists, physician-scientists, advocate leaders, trainees, students and individuals with Ras syndromes and their families. The goals, to discuss basic science and clinical issues, to set forth a solid framework for future research, to direct translational applications towards therapy and to set forth best practices for individuals with RASopathies was successfully meet with a commitment to begin to move towards clinical trials.
- Published
- 2009
- Full Text
- View/download PDF
12. Potent 2-[(pyrimidin-4-yl)amine}-1,3-thiazole-5-carbonitrile-based inhibitors of VEGFR-2 (KDR) kinase
- Author
-
John T. Sisko, Christine Fernandes, Bradley K. Wong, Patrice A. Ciecko, Nancy E. Kohl, Mark T. Bilodeau, Keith W. Rickert, Laura Sepp-Lorenzino, Leonard D. Rodman, Timothy J. Koester, Jackson B. Gibbs, Cynthia Miller-Stein, Debra A. McLoughlin, Joseph J. Lynch, Carolyn A. Buser, George D. Hartman, Xianzhi Mao, Kathleen E. Coll, Kenneth A. Thomas, Jennifer M. Shipman, and Thomas J. Tucker
- Subjects
Stereochemistry ,Clinical Biochemistry ,hERG ,Pharmaceutical Science ,Sensitivity and Specificity ,Biochemistry ,Chemical synthesis ,Structure-Activity Relationship ,chemistry.chemical_compound ,Dogs ,Growth factor receptor ,Nitriles ,parasitic diseases ,Drug Discovery ,Animals ,Thiazole ,Protein Kinase Inhibitors ,neoplasms ,Molecular Biology ,chemistry.chemical_classification ,Molecular Structure ,biology ,Chemistry ,Kinase ,Organic Chemistry ,Macaca mulatta ,Vascular Endothelial Growth Factor Receptor-2 ,Rats ,Thiazoles ,Pyrimidines ,Enzyme ,Enzyme inhibitor ,cardiovascular system ,biology.protein ,Molecular Medicine ,Amine gas treating ,circulatory and respiratory physiology - Abstract
Pyrimidino-thiazolyl carbonitriles were prepared that are potent VEGFR-2 (KDR) kinase inhibitors. The modification of lead structures resulted in 3m which exhibited the best overall profile in KDR inhibitory activity, iv/po pharmacokinetics, and reduced hERG affinity.
- Published
- 2006
- Full Text
- View/download PDF
13. Potent N-(1,3-Thiazol-2-yl)pyridin-2-amine Vascular Endothelial Growth Factor Receptor Tyrosine Kinase Inhibitors with Excellent Pharmacokinetics and Low Affinity for the hERG Ion Channel
- Author
-
Keith W. Rickert, Christine Fernandes, Xianzhi Mao, Kathleen E. Coll, George D. Hartman, Bradley K. Wong, Peter J. Manley, Adrienne E. Balitza, Timothy J. Koester, Mark T. Bilodeau, Debra A. McLoughlin, David C. Heimbrook, Rosemary C. McFall, Sean Yu, William R. Huckle, Jackson B. Gibbs, Jennifer M. Shipman, Kenneth A. Thomas, Raju Subramanian, Joseph J. Lynch, Nancy E. Kohl, Leonard D. Rodman, Cynthia Miller-Stein, Laura Sepp-Lorenzino, and Carolyn Buser-Doepner
- Subjects
Male ,ERG1 Potassium Channel ,Pyridines ,Stereochemistry ,hERG ,Administration, Oral ,Aminopyridines ,Biological Availability ,In Vitro Techniques ,Cell Line ,Rats, Sprague-Dawley ,Electrocardiography ,Mice ,Radioligand Assay ,Dogs ,In vivo ,Drug Discovery ,Animals ,Tissue Distribution ,Phosphorylation ,Lung ,biology ,Kinase ,Chemistry ,Macaca mulatta ,Vascular Endothelial Growth Factor Receptor-2 ,Ether-A-Go-Go Potassium Channels ,In vitro ,Rats ,Thiazoles ,Receptors, Vascular Endothelial Growth Factor ,Potassium Channels, Voltage-Gated ,Enzyme inhibitor ,Microsomes, Liver ,biology.protein ,Molecular Medicine ,Amine gas treating ,Signal transduction ,Tyrosine kinase - Abstract
A series of N-(1,3-thiazol-2-yl)pyridin-2-amine KDR kinase inhibitors have been developed that possess optimal properties. Compounds have been discovered that exhibit excellent in vivo potency. The particular challenges of overcoming hERG binding activity and QTc increases in vivo in addition to achieving good pharmacokinetics have been acomplished by discovering a unique class of amine substituents. These compounds have a favorable kinase selectivity profile that can be accentuated with appropriate substitution.
- Published
- 2004
- Full Text
- View/download PDF
14. A Novel Orally Bioavailable Inhibitor of Kinase Insert Domain-Containing Receptor Induces Antiangiogenic Effects and Prevents Tumor Growth in Vivo
- Author
-
Xianzhi Mao, Elaine Rands, Laura Sepp-Lorenzino, Patrice A. Ciecko, Susan Hill, Jennifer M. Shipman, William F. Hoffman, Brett Connolly, Mark E. Fraley, Stephen C. Beck, Kathleen E. Coll, Kenneth A. Thomas, David C. Heimbrook, Keith W. Rickert, Lenora Davis, Jackson B. Gibbs, Nancy E. Kohl, Joanne Antanavage, and George D. Hartman
- Subjects
Vascular Endothelial Growth Factor A ,Cancer Research ,medicine.medical_specialty ,Angiogenesis ,Fibrosarcoma ,Transplantation, Heterologous ,Administration, Oral ,Biological Availability ,Mice, Nude ,Angiogenesis Inhibitors ,Antineoplastic Agents ,Pharmacology ,Biology ,Mice ,chemistry.chemical_compound ,In vivo ,Cell Line, Tumor ,Internal medicine ,medicine ,Animals ,Humans ,Enzyme Inhibitors ,Phosphorylation ,Kinase activity ,Autophosphorylation ,Kinase insert domain receptor ,Tyrosine phosphorylation ,Vascular Endothelial Growth Factor Receptor-2 ,Vascular endothelial growth factor ,Endocrinology ,Oncology ,chemistry ,Cell Division - Abstract
A strategy for antagonizing vascular endothelial growth factor (VEGF) -induced angiogenesis is to inhibit the kinase activity of its receptor, kinase insert domain-containing receptor (KDR), the first committed and perhaps the last unique step in the VEGF signaling cascade. We synthesized a novel ATP-competitive KDR tyrosine kinase inhibitor that potently suppresses human and mouse KDR activity in enzyme (IC50 = 7.8–19.5 nm) and cell-based assays (IC50 = 8 nm). The compound was bioavailable in vivo, leading to a dose-dependent decrease in basal- and VEGF-stimulated KDR tyrosine phosphorylation in lungs from naïve and tumor-bearing mice (IC50 = 23 nm). Pharmacokinetics and pharmacodynamics guided drug dose selection for antitumor efficacy studies. HT1080 nude mice xenografts were treated orally twice daily with vehicle, or 33 or 133 mg/kg of compound. These doses afforded trough plasma concentrations approximately equal to the IC50 for inhibition of KDR autophosphorylation in vivo for the 33 mg/kg group, and higher than the IC99 for the 133 mg/kg group. Chronic treatment at these doses was well-tolerated and resulted in dose-dependent inhibition of tumor growth, decreased tumor vascularization, decreased proliferation, and enhanced cell death. Antitumor efficacy correlated with inhibition of KDR tyrosine phosphorylation in the tumor, as well as in a surrogate tissue (lung). Pharmacokinetics and pharmacodynamics assessment indicated that the degree of tumor growth inhibition correlated directly with the extent of inhibition of KDR tyrosine phosphorylation in tumor or lung at trough. These observations highlight the need to design antiangiogenic drug regimens to ensure constant target suppression and to take advantage of PD end points to guide dose selection.
- Published
- 2004
- Full Text
- View/download PDF
15. The synthesis and biological evaluation of a series of potent dual inhibitors of farnesyl and geranyl-Geranyl protein transferases
- Author
-
Christine Fernandes, Samuel L. Graham, George D. Hartman, Joseph P. Davide, William C. Lumma, Jackson B. Gibbs, Robert B. Lobell, Hans E. Huber, Carolyn A. Buser, Michelle Ellis-Hutchings, Ronald G. Robinson, Thomas J. Tucker, John T. Sisko, Anthony M Smith, Marc Abrams, and Dongming Liu
- Subjects
Farnesyl Protein Transferase ,Stereochemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Binding, Competitive ,environment and public health ,Biochemistry ,Pyrophosphate ,Inhibitory Concentration 50 ,Structure-Activity Relationship ,chemistry.chemical_compound ,Prenylation ,Drug Discovery ,Tumor Cells, Cultured ,Transferase ,Enzyme Inhibitors ,Molecular Biology ,chemistry.chemical_classification ,Farnesyltranstransferase ,Farnesyl-diphosphate farnesyltransferase ,Alkyl and Aryl Transferases ,Binding Sites ,biology ,Organic Chemistry ,rap GTP-Binding Proteins ,Enzyme ,chemistry ,Enzyme inhibitor ,biology.protein ,Molecular Medicine ,Protein Binding - Abstract
We have prepared a series of potent, dual inhibitors of the prenyl transferases farnesyl protein transferase (FPTase) and geranyl-geranyl protein transferase I (GGPTase). The compounds were shown to possess potent activity against both enzymes in cell culture. Mechanistic analysis has shown that the compounds are CAAX competitive for FPTase inhibition but geranyl-geranyl pyrophosphate (GGPP) competitive for GGPTase inhibiton.
- Published
- 2002
- Full Text
- View/download PDF
16. 3-Aminopyrrolidinone Farnesyltransferase Inhibitors: Design of Macrocyclic Compounds with Improved Pharmacokinetics and Excellent Cell Potency
- Author
-
Kelly Hamilton, Michael J. Bogusky, Robert B. Lobell, Lorena S. Beese, Joel R. Huff, Samuel L. Graham, Marc Abrams, Jackson B. Gibbs, Christine Fernandes, Ronald G. Robinson, J. Christopher Culberson, Carl F. Homnick, Eileen S. Walsh, Joseph J. Lynch, David C. Heimbrook, Michelle Ellis-Hutchings, Douglas C. Beshore, Hans E. Huber, Kenneth S. Koblan, Jeffrey S. Taylor, George D. Hartman, Hema Bhimnathwala, Kelem Kassahun, Nancy E. Kohl, Theresa M. Williams, Carolyn A. Buser, A. David Rodrigues, Joseph P. Davide, C. Blair Zartman, Steven N. Gallicchio, and Ian M. Bell
- Subjects
Models, Molecular ,ERG1 Potassium Channel ,Magnetic Resonance Spectroscopy ,Potassium Channels ,Pyrrolidines ,Stereochemistry ,Farnesyltransferase ,In Vitro Techniques ,Naphthalenes ,Crystallography, X-Ray ,Mass Spectrometry ,Cell Line ,Electrocardiography ,Structure-Activity Relationship ,Dogs ,Transcriptional Regulator ERG ,In vivo ,Drug Discovery ,Animals ,Cytochrome P-450 CYP3A ,Cytochrome P-450 Enzyme Inhibitors ,Farnesyltranstransferase ,Humans ,Structure–activity relationship ,Enzyme Inhibitors ,Cation Transport Proteins ,Farnesyl-diphosphate farnesyltransferase ,Alkyl and Aryl Transferases ,Molecular Structure ,biology ,Chemistry ,Oxidoreductases, N-Demethylating ,Stereoisomerism ,Ether-A-Go-Go Potassium Channels ,Bioavailability ,DNA-Binding Proteins ,Potassium Channels, Voltage-Gated ,Enzyme inhibitor ,Microsomes, Liver ,Trans-Activators ,biology.protein ,Molecular Medicine ,Aryl Hydrocarbon Hydroxylases ,Chromatography, Liquid ,Protein Binding - Abstract
A series of macrocyclic 3-aminopyrrolidinone farnesyltransferase inhibitors (FTIs) has been synthesized. Compared with previously described linear 3-aminopyrrolidinone FTIs such as compound 1, macrocycles such as 49 combined improved pharmacokinetic properties with a reduced potential for side effects. In dogs, oral bioavailability was good to excellent, and increases in plasma half-life were due to attenuated clearance. It was observed that in vivo clearance correlated with the flexibility of the molecules and this concept proved useful in the design of FTIs that exhibited low clearance, such as FTI 78. X-ray crystal structures of compounds 49 and 66 complexed with farnesyltransferase (FTase)-farnesyl diphosphate (FPP) were determined, and they provide details of the key interactions in such ternary complexes. Optimization of this 3-aminopyrrolidinone series of compounds led to significant increases in potency, providing 83 and 85, the most potent inhibitors of FTase in cells described to date.
- Published
- 2002
- Full Text
- View/download PDF
17. Design and Biological Activity of (S)-4-(5-{[1-(3-Chlorobenzyl)-2- oxopyrrolidin-3-ylamino]methyl}imidazol-1-ylmethyl)benzonitrile, a 3-Aminopyrrolidinone Farnesyltransferase Inhibitor with Excellent Cell Potency
- Author
-
Theresa M. Williams, A. David Rodrigues, Douglas C. Beshore, Carolyn A. Buser, Joseph P. Davide, Michelle Ellis-Hutchings, Steven N. Gallicchio, J. Christopher Culberson, Samuel L. Graham, George D. Hartman, Marc Abrams, David C. Heimbrook, and Eileen S. Walsh, Carl F. Homnick, Kenneth S. Koblan, Joel R. Huff, Robert B. Lobell, Joseph J. Lynch, Christine Fernandes, Patricia A. Miller, Jackson B. Gibbs, Charles A. Omer, Kelem Kassahun, Nancy E. Kohl, and Ian M. Bell
- Subjects
Models, Molecular ,Lactams ,Stereochemistry ,Farnesyltransferase ,Biological Availability ,Antineoplastic Agents ,Mice, Transgenic ,Binding, Competitive ,Mice ,Radioligand Assay ,Structure-Activity Relationship ,Dogs ,Nitriles ,Drug Discovery ,Animals ,Farnesyltranstransferase ,Potency ,Structure–activity relationship ,Enzyme Inhibitors ,Cell potency ,Cell Line, Transformed ,Farnesyl-diphosphate farnesyltransferase ,Alkyl and Aryl Transferases ,Binding Sites ,biology ,Chemistry ,Farnesyltransferase inhibitor ,Imidazoles ,Stereoisomerism ,Biological activity ,Neoplasms, Experimental ,Pyrrolidinones ,Genes, ras ,Enzyme inhibitor ,Drug Design ,biology.protein ,Molecular Medicine ,Drug Screening Assays, Antitumor - Abstract
The synthesis, structure-activity relationships, and biological properties of a novel series of imidazole-containing inhibitors of farnesyltransferase are described. Starting from a 3-aminopyrrolidinone core, a systematic series of modifications provided 5h, a non-thiol, non-peptide farnesyltransferase inhibitor with excellent bioavailability in dogs. Compound 5h was found to have an unusually favorable ratio of cell potency to intrinsic potency, compared with other known FTIs. It exhibited excellent potency against a range of tumor cell lines in vitro and showed full efficacy in the K-rasB transgenic mouse model.
- Published
- 2001
- Full Text
- View/download PDF
18. Oxo-piperazine Derivatives of N-Arylpiperazinones as Inhibitors of Farnesyltransferase
- Author
-
I.-W. Chen, Kenneth S. Koblan, George D. Hartman, Robert B. Lobell, Theresa M. Williams, Samuel L. Graham, Jackson B. Gibbs, Ian B Greenberg, Debra A. McLoughlin, Dongming Liu, Jeffrey M. Bergman, Timothy V. Olah, C. Blair Zartman, Donna Wei, Timothy J. O'Neill, Joseph P. Davide, Nancy E. Kohl, and Christopher J. Dinsmore
- Subjects
Stereochemistry ,Farnesyltransferase ,Clinical Biochemistry ,Pharmaceutical Science ,Biochemistry ,Chemical synthesis ,Piperazines ,Structure-Activity Relationship ,chemistry.chemical_compound ,Dogs ,Drug Discovery ,Animals ,Farnesyltranstransferase ,Structure–activity relationship ,Potency ,Enzyme Inhibitors ,Molecular Biology ,Farnesyl-diphosphate farnesyltransferase ,Alkyl and Aryl Transferases ,biology ,Organic Chemistry ,Piperazine ,chemistry ,Enzyme inhibitor ,biology.protein ,Lactam ,Molecular Medicine - Abstract
The evaluation of SAR associated with the insertion of carbonyl groups at various positions of N-arylpiperazinone farnesyltransferase inhibitors is described herein. 1-Aryl-2,3-diketopiperazine derivatives exhibited the best balance of potency and pharmacokinetic profile relative to the parent 1-aryl-2-piperazinones.
- Published
- 2001
- Full Text
- View/download PDF
19. Farnesyltransferase Inhibitors Potentiate the Antitumor Effect of Radiation on a Human Tumor Xenograft Expressing Activated HRAS1
- Author
-
George J. Cerniglia, Donna Kusewitt, Said M. Sebti, Oliff Allen I, Nancy E. Kohl, Rosemarie Mick, Eric J. Bernhard, Jackson B. Gibbs, Andrew D. Hamilton, Sydney M. Evans, W. Gillies McKenna, Ruth J. Muschel, and Elizabeth Cohen-Jonathan
- Subjects
Farnesyltranstransferase ,Radiation ,biology ,Ratón ,Farnesyltransferase ,Biophysics ,In vitro ,Tissue culture ,In vivo ,Immunology ,Cancer research ,biology.protein ,Radiology, Nuclear Medicine and imaging ,HRAS ,Radiosensitivity - Abstract
Successful radiosensitization requires that tumor cells become more radiosensitive without causing an equivalent reduction in the survival of cells of the surrounding normal tissues. Since tumor cell radiosensitivity can be influenced by RAS oncogene activation, we have hypothesized that inhibition of oncogenic RAS activity would lead to radiosensitization of tumors with activated RAS. We previously showed in tissue culture that prenyltransferase treatment of cells with activated RAS resulted in radiosensitization, whereas treatment of cells with wild-type RAS had no effect on radiation survival. Here we ask whether the findings obtained in vitro have applicability in vivo. We found that treatment of nude mice bearing T24 tumor cell xenografts with farnesyltransferase inhibitors resulted in a significant and synergistic reduction in tumor cell survival after irradiation. The regrowth of T24 tumors expressing activated RAS was also significantly prolonged by the addition of treatment with farnesyltransferase inhibitors compared to the regrowth after irradiation alone. In contrast, there was no effect on the radiosensitivity of HT-29 tumors expressing wild-type RAS. These results demonstrate that specific radiosensitization of tumors expressing activated RAS oncogenes can be obtained in vivo.
- Published
- 2000
- Full Text
- View/download PDF
20. Mechanism-Based Target Identification and Drug Discovery in Cancer Research
- Author
-
Jackson B. Gibbs
- Subjects
medicine.medical_specialty ,Neoplasms, Hormone-Dependent ,Chemistry, Pharmaceutical ,Population ,Mechanism based ,Antineoplastic Agents ,Breast Neoplasms ,Disease ,Biology ,Neoplasms ,medicine ,Humans ,Enzyme Inhibitors ,Intensive care medicine ,education ,Clinical Trials as Topic ,education.field_of_study ,Multidisciplinary ,Drug discovery ,Cancer ,Genetic Therapy ,medicine.disease ,Drug development ,Drug Design ,Identification (biology) ,Drug Screening Assays, Antitumor ,Human cancer - Abstract
Cancer as a disease in the human population is becoming a larger health problem, and the medicines used as treatments have clear limitations. In the past 20 years, there has been a tremendous increase in our knowledge of the molecular mechanisms and pathophysiology of human cancer. Many of these mechanisms have been exploited as new targets for drug development in the hope that they will have greater antitumor activity with less toxicity to the patient than is seen with currently used medicines. The fruition of these efforts in the clinic is just now being realized with a few encouraging results.
- Published
- 2000
- Full Text
- View/download PDF
21. In Vitro and In Vivo Effects of a Farnesyltransferase Inhibitor onNf1-Deficient Hematopoietic Cells
- Author
-
Brigit R. Taylor, Nidal Mahgoub, Kevin Shannon, Nancy E. Kohl, Jackson B. Gibbs, Tyler Jacks, and Mary Gratiot
- Subjects
Adoptive cell transfer ,Myeloid ,GTPase-activating protein ,biology ,Kinase ,Farnesyltransferase ,Immunology ,Farnesyltransferase inhibitor ,Myeloid leukemia ,Cell Biology ,Hematology ,Biochemistry ,medicine.anatomical_structure ,Prenylation ,medicine ,Cancer research ,biology.protein - Abstract
Oncogenic RAS alleles encode proteins that accumulate in the guanosine triphosphate (GTP)-bound state. Because post-translational processing of Ras by farnesyltransferase is essential for biologic function, inhibitors of this enzyme have been developed as rational cancer therapeutics. We have investigated farnesyltransferase inhibitor (FTI) L-744,832 in an in vivo murine model of myeloid leukemia that is associated with inactivation of the Nf1 tumor suppressor gene.Nf1 encodes a GTPase activating protein for Ras, andNf1-deficient (Nf1−/−) hematopoietic cells show hyperactive Ras signaling through the mitogen-activated protein (MAP) kinase pathway. L-744,832 inhibited H-Ras prenylation in cell lines and in primary hematopoietic cells and abrogated the in vitro growth of myeloid progenitor colonies in response to granulocyte-macrophage colony-stimulating factor (GM-CSF). This FTI also partially blocked GM-CSF–induced MAP kinase activation, but did not reduce constitutively elevated levels of MAP kinase activity in primaryNf1−/− cells. Injection of a single dose of 40 or 80 mg/kg of L-744,832 increased the amount of unprocessed H-Ras in bone marrow cells, but had no detectable effect on N-Ras. Adoptive transfer ofNf1−/− hematopoietic cells into irradiated mice induces a myeloproliferative disorder that did not respond to L-744,832 treatment. We speculate that the lack of efficacy in this model is due to the resistance of N-Ras and K-Ras processing to inhibition by this FTI.
- Published
- 1999
- Full Text
- View/download PDF
22. Non-thiol 3-aminomethylbenzamide inhibitors of farnesyl-protein transferase
- Author
-
Kenneth S. Koblan, Christopher J. Dinsmore, Jackson B. Gibbs, S. C. Mactough, J. Christopher Culberson, Terrence M. Ciccarone, Nancy E. Kohl, Samuel L. Graham, Daksha Shah, Timothy J. O'Neill, Oliff Allen I, Theresa M. Williams, and George D. Hartman
- Subjects
Models, Molecular ,Farnesyl Protein Transferase ,Protein Conformation ,Stereochemistry ,Clinical Biochemistry ,Drug Evaluation, Preclinical ,Pharmaceutical Science ,Biochemistry ,Inhibitory Concentration 50 ,Structure-Activity Relationship ,Drug Discovery ,Animals ,Structure–activity relationship ,Transferase ,Sulfhydryl Compounds ,Enzyme Inhibitors ,Binding site ,Molecular Biology ,chemistry.chemical_classification ,Farnesyltranstransferase ,Alkyl and Aryl Transferases ,Binding Sites ,Molecular Structure ,biology ,Chemistry ,Organic Chemistry ,Imidazoles ,Rats ,Enzyme ,Enzyme inhibitor ,Drug Design ,Benzamides ,biology.protein ,Thiol ,Molecular Medicine ,Cell Division - Abstract
The design and syntheses of non-thiol inhibitors of farnesyl-protein transferase are described. Substitutions on an imidazolylmethyl-AMBA-methionine template gave a highly potent and cell-active inhibitor.
- Published
- 1999
- Full Text
- View/download PDF
23. Clavaric Acid and Steroidal Analogues as Ras- and FPP-Directed Inhibitors of Human Farnesyl-Protein Transferase
- Author
-
Russell B. Lingham, Jackson B. Gibbs, James D. Bergstrom, Rew Dj, Francine R. Wilson, Kim Bm, Samuel L. Graham, Hiranthi Jayasuriya, Kenneth S. Koblan, Sheo B. Singh, Schaber, Amo Se, Keith C. Silverman, and Nancy E. Kohl
- Subjects
Farnesyl-diphosphate farnesyltransferase ,Farnesyl Protein Transferase ,biology ,Squalene synthase activity ,Reductase ,biology.organism_classification ,chemistry.chemical_compound ,Biochemistry ,chemistry ,Enzyme inhibitor ,Drug Discovery ,Clavariadelphus truncatus ,biology.protein ,Molecular Medicine ,Protein prenylation ,Clavaric acid - Abstract
We have identified a novel fungal metabolite that is an inhibitor of human farnesyl-protein transferase (FPTase) by randomly screening natural product extracts using a high-throughput biochemical assay. Clavaric acid [24, 25-dihydroxy-2-(3-hydroxy-3-methylglutaryl)lanostan-3-one] was isolated from Clavariadelphus truncatus; it specifically inhibits human FPTase (IC50 = 1.3 microM) and does not inhibit geranylgeranyl-protein transferase-I (GGPTase-I) or squalene synthase activity. It is competitive with respect to Ras and is a reversible inhibitor of FPTase. An alkaline hydrolysis product of clavaric acid, clavarinone [2,24,25-trihydroxylanostan-3-one], lacking the 3-hydroxy-3-methylglutaric acid side chain is less active as a FPTase inhibitor. Similarly, a methyl ester derivative of clavaric acid is also inactive. In Rat1 ras-transformed cells clavaric acid and lovastatin inhibited Ras processing without being overtly cytotoxic. Excess mevalonate reversed the effects of lovastatin but not of clavaric acid suggesting that the block on Ras processing by clavaric acid was due to inhibition of FPTase and not due to inhibition of HMG-CoA reductase. Despite these results, the possibility existed that clavaric acid inhibited Ras processing by directly inhibiting HMG-CoA reductase. To directly examine the effects of clavaric acid and clavarinone on HMG-CoA reductase, cholesterol synthesis was measured in HepG2 cells. No inhibition of HMG-CoA reductase was observed indicating that the inhibition of Ras processing by this class of compounds is due to inhibition of FPTase. To date, clavaric acid is the second reported nitrogen-free compound that competes with Ras to inhibit FPTase activity. A series of related compounds derived from computer-based similarity searches and subsequent rational chemical synthetic design provided compounds that exhibited a range of activity (0.04 --> 100 microM) against FPTase. Modest changes in the structures of these inhibitors dramatically change the inhibitory activity of these inhibitors.
- Published
- 1998
- Full Text
- View/download PDF
24. N-Arylalkyl Pseudopeptide Inhibitors of Farnesyltransferase
- Author
-
S J deSolms, Rands E, Jackson B. Gibbs, Catherine M. Wiscount, Scholz Th, Robert L. Smith, Kenneth S. Koblan, Nancy E. Kohl, E A Giuliani, David L. Pompliano, Scott D. Mosser, Oliff Allen I, and Samuel L. Graham
- Subjects
Stereochemistry ,Farnesyltransferase ,Naphthalenes ,Oncogene Protein p21(ras) ,Chemical synthesis ,Mice ,Structure-Activity Relationship ,Prenylation ,Drug Discovery ,Animals ,Farnesyltranstransferase ,Structure–activity relationship ,Prodrugs ,Enzyme Inhibitors ,Cell Line, Transformed ,chemistry.chemical_classification ,Farnesyl-diphosphate farnesyltransferase ,Alkyl and Aryl Transferases ,biology ,Tetrapeptide ,Chemistry ,Molecular Mimicry ,Esters ,3T3 Cells ,Rats ,Enzyme ,Enzyme inhibitor ,biology.protein ,Molecular Medicine ,Oligopeptides ,Protein Processing, Post-Translational ,Cell Division - Abstract
Inhibitors of Ras protein farnesyltransferase are described which are reduced pseudopeptides related to the C-terminal tetrapeptide of the Ras protein that signals farnesylation. Reduction of the carbonyl groups linking the first three residues of the tetrapeptide leads to active inhibitors which are chemically unstable. Stability can be restored by alkylating the central amine of the tetrapeptide. Studies of the SAR of these alkylated pseudopeptides with concomitant modification of the side chain of the third residue led to 2(S)-(2(S)-¿[2(S)-(2(R)-amino-3-mercaptopropylamino)-3(S)- methylpentyl]naphthalen-1-ylmethylamino¿acetylamino)-4 -methylsulfany lbutyric acid (11), a subnanomolar inhibitor. The methyl ester (10) of this compound exhibited submicromolar activity in the processing assay and selectively inhibited anchorage-independent growth of Rat1 cells transformed by v-ras at 2.5-5 microM.
- Published
- 1998
- Full Text
- View/download PDF
25. DIARYLETHER INHIBITORS OF FARNESYL-PROTEIN TRANSFERASE
- Author
-
Theresa M. Williams, Rands E, Nancy E. Kohl, George D. Hartman, Samuel L. Graham, Kelly Hamilton, Timothy J. O'Neill, Oliff Allen I, Christopher J. Dinsmore, Kenneth S. Koblan, and Jackson B. Gibbs
- Subjects
chemistry.chemical_classification ,Farnesyl-diphosphate farnesyltransferase ,biology ,Farnesyl Protein Transferase ,Tetrapeptide ,medicine.drug_class ,Stereochemistry ,Organic Chemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Carboxamide ,Biochemistry ,Chemical synthesis ,Enzyme ,chemistry ,Enzyme inhibitor ,Drug Discovery ,medicine ,biology.protein ,Molecular Medicine ,Transferase ,Molecular Biology - Abstract
The design and synthesis of simple nonpeptide inhibitors of farnesyl-protein transferase (FTase) are described. Cysteine-derived diarylether frameworks are appropriate structural replacements for the C -terminal tetrapeptide portion of the Ras protein, and possess in vitro potency against FTase. Inhibitory activity is dependent on the ring-substitution pattern, and does not require the presence of a C -terminal carboxylate group. © 1997 Elsevier Science Ltd.
- Published
- 1997
- Full Text
- View/download PDF
26. THE POTENTIAL OF FARNESYLTRANSFERASE INHIBITORS AS CANCER CHEMOTHERAPEUTICS
- Author
-
Oliff Allen I and Jackson B. Gibbs
- Subjects
Farnesyltransferase ,Protein Prenylation ,Antineoplastic Agents ,Toxicology ,Mice ,Geranylgeranylation ,Prenylation ,Transferases ,Anti-apoptotic Ras signalling cascade ,Genes, Regulator ,Tumor Cells, Cultured ,medicine ,Animals ,Farnesyltranstransferase ,Humans ,Enzyme Inhibitors ,Pharmacology ,Alkyl and Aryl Transferases ,biology ,Cancer ,Neoplasms, Experimental ,Subcellular localization ,medicine.disease ,Cell biology ,Cell Transformation, Neoplastic ,Genes, ras ,biology.protein ,Signal transduction - Abstract
Mutant ras oncogenes and alterations in the mitogenic signaling pathways that they regulate are associated with a wide variety of solid tumors and leukemias for which existing chemotherapeutics have limited utility. Of the possible approaches to inhibit Ras function, much attention has focused on a posttranslational modification, farnesylation, which is required for the subcellular localization of Ras to the plasma membrane and is critical to Ras cell-transforming activity. Inhibitors of the enzyme that catalyzes Ras farnesylation, farnesyl-protein transferase (FPTase), have been developed. These compounds inhibit the tumorigenic phenotypes of ras-transformed cells and human tumor cells in cell culture and in animal models. Moreover, FPTase inhibitors have not demonstrated toxicity to normal cells in culture or to normal tissues in mice. FPTase inhibitors are among the first small molecule compounds designed from studies of oncogenes that might serve as novel cancer chemotherapeutics.
- Published
- 1997
- Full Text
- View/download PDF
27. Farnesyl: proteintransferase inhibitors as agents to inhibit tumor growth
- Author
-
Ch. J. Dinsmore, George D. Hartman, Ch. A. Omer, Neville J. Anthony, Kenneth S. Koblan, S J deSolms, Jackson B. Gibbs, A. L. Burkhardt, Robert B. Lobell, Nancy E. Kohl, Oliff Allen I, C. A. Buser‐Doepner, and Theresa M. Williams
- Subjects
Clinical Biochemistry ,Protein Prenylation ,Antineoplastic Agents ,Biology ,Biochemistry ,Prenylation ,Transferases ,Neoplasms ,Tumor Cells, Cultured ,Animals ,Humans ,Tumor growth ,Enzyme Inhibitors ,chemistry.chemical_classification ,Alkyl and Aryl Transferases ,General Medicine ,Cell biology ,Genes, ras ,Membrane ,Enzyme ,chemistry ,ras Proteins ,Molecular Medicine ,Tumor growth inhibition ,lipids (amino acids, peptides, and proteins) - Abstract
Ras, a signal-transducing protein involved in mediating growth factor-stimulated proliferation, is mutationally activated in over 30% of human tumors. To be functional Ras must bind to the inner surface of the plasma membrane, with post-translational lipid modifications being necessary for this localization. The essential, first modification of Ras is farnesylation catalyzed by the enzyme farnesyl: proteintransferase (FPTase). Inhibitors of FPTase (FTIs) are currently being tested to determine if they are capable of tumor growth inhibition. Here we describe our efforts, along with those of other groups, in testing the biological and biochemical effects of FTIs.
- Published
- 1997
- Full Text
- View/download PDF
28. Selection of Potent Inhibitors of Farnesyl-protein Transferase from a Synthetic Tetrapeptide Combinatorial Library
- Author
-
Jackson B. Gibbs, Patricia Miller, Scott D. Mosser, Dorothy J. Marquis-Omer, Riccardo Cortese, Michael D. Schaber, Kenneth S. Koblan, Andrew L. Wallace, Antonello Pessi, Kelly Hamilton, Charles A. Omer, and Oliff Allen I
- Subjects
chemistry.chemical_classification ,Alkyl and Aryl Transferases ,Farnesyl Protein Transferase ,Tetrapeptide ,Peptidomimetic ,Stereochemistry ,Circular Dichroism ,Antineoplastic Agents ,Peptide ,Cell Biology ,Ligand (biochemistry) ,Biochemistry ,Amino acid ,chemistry.chemical_compound ,chemistry ,Transferases ,Carboxyglutamic acid ,Transferase ,Oligopeptides ,Molecular Biology ,Gene Library - Abstract
Inhibitors of farnesyl-protein transferase (FPTase) show promise as anticancer agents. Based on the sequence of the protein substrates of FPTase (the CAAX sequence), potent and selective peptidomimetic inhibitors have been developed; these compounds share with the peptide substrate a free thiol and a C-terminal carboxylate. We have used a synthetic tetrapeptide combinatorial library to screen for new leads devoid of these features: the peptides were C-terminally amidated, and no free thiol was included in the combinatorial building blocks. To compensate for this negative bias, an expanded set of 68 amino acids was used, including both L and D as well as many non-coded residues. Sixteen individual tetrapeptides derived from the consensus were synthesized and tested; all were active, showing IC50 values ranging from low micromolar to low nanomolar. The most active peptide, D-tryptophan-D-methionine-D-4-chlorophenylalanine-L-gamma- carboxyglutamic acid (Ki = 2 nM), is also very selective showing little inhibitory activity against the related enzyme geranylgeranyl-protein transferase type I (IC50 > 50 microM). In contrast to CAAX-based peptidomimetics, D-tryptophan-D-methionine-D-4-chlorophenylalanine-L-gamma-carboxyglut amic acid appeared to mimic the isoprenoid substrate farnesyl diphosphate as determined by kinetic and physical measurements. D-Tryptophan-Dmethionine-D-4-chlorophenylalanine-L-gamma- carboxyglutamic acid was a competitive inhibitor of FPTase with respect to farnesyl diphosphate substrate and uncompetitive with respect to CAAX substrate. Furthermore, we demonstrated that FPTase undergoes ligand dependent conformational changes in its circular dichroism spectrum and that D-tryptophan-D-methionine-D-4-chlorophenylalanine-L-gamma- carboxyglutamic acid induced a conformational change identical to that observed with farnesyl diphosphate ligand.
- Published
- 1996
- Full Text
- View/download PDF
29. New Molecular Targets for Cancer Therapy
- Author
-
Oliff Allen I, Jackson B. Gibbs, and Frank McCormick
- Subjects
Multidisciplinary ,DNA Repair ,business.industry ,Genetic Vectors ,Gene Transfer Techniques ,Cancer therapy ,Genetic Therapy ,Oncogenes ,Computational biology ,Gene Expression Regulation, Neoplastic ,Text mining ,Neoplasms ,Viruses ,Molecular targets ,Humans ,Medicine ,Genes, Tumor Suppressor ,business - Published
- 1996
- Full Text
- View/download PDF
30. Farnesyltransferase inhibitors and anti-Ras therapy
- Author
-
Jackson B. Gibbs, Oliff Allen I, S. Jane Desolms, Laura Sepp-Lorenzino, Kenneth S. Koblan, Nancy E. Kohl, Neal Rosen, Michael W. Conner, Samuel L. Graham, Theresa M. Williams, Charles A. Omer, George D. Hartman, and Neville J. Anthony
- Subjects
Cancer Research ,Farnesyl-diphosphate farnesyltransferase ,Alkyl and Aryl Transferases ,biology ,Farnesyltransferase ,Cell ,Antineoplastic Agents ,Biological activity ,environment and public health ,medicine.anatomical_structure ,Oncology ,Biochemistry ,Prenylation ,Transferases ,Cell culture ,Mitogen-activated protein kinase ,ras Proteins ,biology.protein ,medicine ,Animals ,Farnesyltranstransferase ,Humans ,Guanosine Triphosphate ,Enzyme Inhibitors ,Signal transduction - Abstract
The oncoprotein encoded by mutant ras genes is initially synthesized as a cytoplasmic precursor which requires posttranslational processing to attain biological activity; farnesylation of the cysteine residue present in the CaaX motif located at the carboxy-terminus of all Ras proteins is the critical modification. Once farnesylated and further modified, the mature Ras protein is inserted into the cell's plasma membrane where it participates in the signal transduction pathways that control cell growth and differentiation. The farnesylation reaction that modifies Ras and other cellular proteins having an appropriate CaaX motif is catalyzed by a housekeeping enzyme termed farnesyl-protein transferase (FPTase). Inhibitors of this enzyme have been prepared by several laboratories in an effort to identify compounds that would block Ras-induced cell transformation and thereby function as Ras-specific anticancer agents. A variety of natural products and synthetic organic compounds were found to block farnesylation of Ras proteins in vitro. Some of these compounds exhibit antiproliferative activity in cell culture, block the morphological alterations associated with Ras-transformation, and can block the growth of Ras-transformed cell lines in tumor colony-forming assays. By contrast, these compounds do not affect the growth or morphology of cells transformed by the Raf or Mos oncoproteins, which do not require farnesylation to achieve biological activity. The efficacy and lack of toxicity observed with FPTase inhibitors in an animal tumor model suggest that specific FPTase inhibitors may be useful for the treatment of some types of cancer.
- Published
- 1996
- Full Text
- View/download PDF
31. New fungal metabolites as potential antihypercholesterolemics and anticancer agents
- Author
-
Gerald F. Bills, Marina Mojena, Keith C. Silverman, Leeyuan Huang, Claude Dufresne, Maria S. Meinz, Manuel Sanchez, Jackson B. Gibbs, James D. Bergstrom, John D. Karkas, Sheo B. Singh, Mary Nallin-Omstead, Russell B. Lingham, Guy H. Harris, and Wendy H. Clapp
- Subjects
biology ,ATP synthase ,Trichoderma viride ,Zaragozic acid ,macromolecular substances ,Plant Science ,Fungus ,biology.organism_classification ,Sporormiella intermedia ,chemistry.chemical_compound ,Squalene ,chemistry ,Biochemistry ,biology.protein ,Transferase ,Fusidium griseum - Abstract
Several potent inhibitors of squalence synthetase have been discovered. Zaragozic acid A is produced by several fungi; zaragozic acid B is produced by several strains of Sporormiella intermedia; zaragozic acids C, E, and F are produced by Leptodontidium elatius; zaragozic acids D and D2 are produced by Amauroascus niger. L-731,120 and L-731,128 are minor components and coproduced with zaragozic acids A and B, respectively. Viridiofungins A, B, and C are produced by Trichoderma viride. Viridiofungin A is also produced by an unidentified sterile fungus. Several of the zaragozic acids are also potent inhibitors of farnesyl-protein transferase (FPTase). Inhibitors of FPTase may act as potential anticancer drugs. Chaetomellic acids A and B are produced by a fungus, Chaetomella acutiseta, while fusidienol is produced by Fusidium griseum. All three compounds are potent inhibitors of FPTase. Our experiences suggest that many novel inhibitors of both squalene synthase and FPTase are produced within a diverse phylogenetic array of filamentous fungi. Several of the zaragozic acids are potent inhibitors of both FPTase and squalene synthases. This is consistent with our observations that zaragozic acids and chaetomellic acids share some structural similarity. Key words: natural inhibitors, squalene synthase, farnesyl-protein transferase.
- Published
- 1995
- Full Text
- View/download PDF
32. Actinoplanic acids A and B as novel inhibitors of farnesyl-protein transferase
- Author
-
Janet M. Sigmund, Isabel Martin, Olga Genilloud, Russell B. Lingham, Magda M. Gagliardi, Brian Heimbuch, Jackson B. Gibbs, Suzanne E. Gartner, Mary Nallin-Omstead, Gregory E. Koch, Sheo B. Singh, George M. Garrity, Carmen Cascales, Manuel Sanchez, Maria Teresa Diez, Charles F. Hirsch, and Keith C. Silverman
- Subjects
chemistry.chemical_classification ,Alkyl and Aryl Transferases ,biology ,Farnesyl Protein Transferase ,Streptomycetaceae ,Stereochemistry ,Carboxylic acid ,General Medicine ,biology.organism_classification ,Applied Microbiology and Biotechnology ,Streptomyces ,Lactones ,Enzyme ,chemistry ,Biochemistry ,Transferases ,Actinomycetales ,Farnesyltranstransferase ,Transferase ,lipids (amino acids, peptides, and proteins) ,Enzyme Inhibitors ,Actinoplanes ,Biotechnology - Abstract
Actinoplanic acids A and B are macrocyclic polycarboxylic acids that are potent reversible inhibitors of farnesyl-protein transferase. Actinoplanic acids A and B were isolated from Actinoplanes sp. MA 7066 while actinoplanic acid B was isolated from both MA 7066 and Streptomyces sp. MA 7099. Actinoplanic acids A and B are competitive with respect to farnesyl diphosphate and are selective inhibitors of farnesyl-protein transferase because they do not inhibit geranylgeranyl-protein transferase type 1 or squalene synthase. MA 7066 is believed to be a novel species of actinomycetes while MA 7099 is believed to be a novel strain of Streptomyces violaceusniger on the basis of morphological, biochemical and chemotaxonomic characteristics as well as its production of actinoplanic acids.
- Published
- 1995
- Full Text
- View/download PDF
33. Inhibition of farnesyltransferase induces regression of mammary and salivary carcinomas in ras transgenic mice
- Author
-
Nancy E. Kohl, Charles A. Omer, Michael W. Conner, Neville J. Anthony, Joseph P. Davide, S. Jane Desolms, Elizabeth A. Giuliani, Robert P. Gomez, Samuel L. Graham, Kelly Hamilton, Laurence K. Handt, George D. Hartman, Kenneth S. Koblan, Astrid M. Kral, Patricia J. Miller, Scott D. Mosser, Timothy J. O'Neill, Elaine Rands, Michael D. Schaber, Jackson B. Gibbs, and Allen Oliff
- Subjects
Genetically modified mouse ,medicine.medical_specialty ,Farnesyl Protein Transferase ,Transgene ,Farnesyltransferase ,Antineoplastic Agents ,Mice, Transgenic ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Methionine ,Transferases ,Internal medicine ,medicine ,Animals ,Enzyme Inhibitors ,chemistry.chemical_classification ,Farnesyltranstransferase ,Alkyl and Aryl Transferases ,Dose-Response Relationship, Drug ,Farnesyl Transferase Inhibitor ,Farnesyltransferase inhibitor ,Mammary Neoplasms, Experimental ,General Medicine ,Salivary Gland Neoplasms ,Genes, ras ,Enzyme ,Endocrinology ,chemistry ,Cancer research ,biology.protein ,Female - Abstract
For Ras oncoproteins to transform mammalian cells, they must be post-translationally modified with a farnesyl group in a reaction catalysed by the enzyme farnesyl-protein transferase (FPTase). Inhibitors of FPTase have therefore been proposed as anti-cancer agents. We show that L-744,832, which mimics the CaaX motif to which the farnesyl group is added, is a potent and selective inhibitor of FPTase. In MMTV-v-Ha-ras mice bearing palpable tumours, daily administration of L-744,832 caused tumour regression. Following cessation of treatment, tumours reappeared, the majority of which regressed upon retreatment. No systemic toxicity was found upon necropsy of L-744,832-treated mice. This first demonstration of anti-FPTase-mediated tumour regression suggests that FPTase inhibitors may be safe and effective anti-tumour agents in some cancers.
- Published
- 1995
- Full Text
- View/download PDF
34. Pharmaceutical research in molecular oncology
- Author
-
Oliff Allen I and Jackson B. Gibbs
- Subjects
Drug Industry ,Protein Conformation ,Receptor Protein-Tyrosine Kinases ,Computational biology ,Biology ,Second Messenger Systems ,Molecular oncology ,General Biochemistry, Genetics and Molecular Biology ,Drug Design ,Neoplasms ,Humans ,Receptors, Growth Factor ,Pharmaceutical sciences ,Protein Kinases ,Cell Division ,Protein Binding ,Signal Transduction - Published
- 1994
- Full Text
- View/download PDF
35. Synthesis and biological activity of ras farnesyl protein transferase inhibitors. Tetrapeptide analogs with amino methyl and carbon linkages
- Author
-
Dona L. Bamberger, Jackson B. Gibbs, Oliff Allen I, Nancy E. Kohl, Samuel L. Graham, Rands E, Thorsten E. Fisher, Robert L. Smith, Scott D. Mosser, John S. Wai, and David L. Pompliano
- Subjects
Farnesyl Protein Transferase ,Stereochemistry ,Molecular Sequence Data ,Clinical Biochemistry ,Pharmaceutical Science ,chemistry.chemical_element ,Biochemistry ,3T3 cells ,Mice ,Structure-Activity Relationship ,chemistry.chemical_compound ,Transferases ,Drug Discovery ,Ic50 values ,medicine ,Animals ,Potency ,Amino Acid Sequence ,Methylene ,Molecular Biology ,Alkyl and Aryl Transferases ,Molecular Structure ,Tetrapeptide ,Chemistry ,Organic Chemistry ,Stereoisomerism ,Biological activity ,3T3 Cells ,Cell Transformation, Viral ,Genes, ras ,medicine.anatomical_structure ,Molecular Medicine ,Oligopeptides ,Carbon - Abstract
Replacement of the central amino methylene linkage of C[psi CH2NH]A[psi CH2NH]AX tetrapeptide inhibitors with carbon tethers led to compounds with potency in the nanomolar range. Some of the more potent olefinic compounds inhibit Ras processing in intact v-ras transformed NIH 3T3 cells with IC50 values in the 0.1 to 1 microM range, and inhibit selectively the anchorage-independent growth of H-ras transformed Rat1 cells at 10 microM.
- Published
- 1994
- Full Text
- View/download PDF
36. Fusidienol: A novel inhibitor of Ras farnesyl-protein transferase from Fusidium griseum
- Author
-
E. Tracy Turner Jones, Sheo B. Singh, Jackson B. Gibbs, Mary Nallin-Omstead, Rosalind G. Jenkins, Russell B. Lingham, Gerald F. Bills, Michael Goetz, and Keith C. Silverman
- Subjects
chemistry.chemical_classification ,Farnesyl Protein Transferase ,Stereochemistry ,Organic Chemistry ,Cancer ,medicine.disease ,Biochemistry ,chemistry ,Drug Discovery ,medicine ,Fusidienol ,Fusidium griseum ,Tricyclic - Abstract
Ras (p21) protein is frequently found mutated in human cancers and must be farnesylated by farnesyl protein-transferase (FPTase) to achieve cell-transforming activity. Our continued search for inhibitors of FPTase as potential cancer chemotherapeutics led to the isolation of fusidienol from extracts of the fungus Fusidium griseum . Fusidienol is a novel and potent oxygen-containing [7/6/6] tricyclic heterocycle.
- Published
- 1994
- Full Text
- View/download PDF
37. Farnesyltransferase inhibitors: Ras research yields a potential cancer therapeutic
- Author
-
Oliff Allen I, Jackson B. Gibbs, and Nancy E. Kohl
- Subjects
chemistry.chemical_classification ,Genetics ,Farnesyl-diphosphate farnesyltransferase ,Alkyl and Aryl Transferases ,biology ,Farnesyl Protein Transferase ,Farnesyltransferase ,Molecular Sequence Data ,Protein Prenylation ,Cancer ,Oncogene Protein p21(ras) ,medicine.disease ,General Biochemistry, Genetics and Molecular Biology ,Cell Transformation, Neoplastic ,Enzyme ,chemistry ,Transferases ,Enzyme inhibitor ,Neoplasms ,biology.protein ,Cancer research ,medicine ,Animals ,Humans ,Amino Acid Sequence - Published
- 1994
- Full Text
- View/download PDF
38. cDNA cloning and expression of rat and human protein geranylgeranyltransferase type-I
- Author
-
C A Omer, Patrick J. Casey, Bruno Giros, Fang L. Zhang, Jackson B. Gibbs, Nancy E. Kohl, and R E Diehl
- Subjects
Protein subunit ,Cell Biology ,Biology ,Biochemistry ,Molecular biology ,Gamma-aminobutyric acid receptor subunit alpha-1 ,Interleukin 10 receptor, alpha subunit ,SCN3A ,Complementary DNA ,Protein geranylgeranyltransferase type I ,Molecular Biology ,Peptide sequence ,G alpha subunit - Abstract
Protein geranylgeranyltransferase type-I (GGTase-I) transfers a geranylgeranyl group to the cysteine residue of candidate proteins containing a carboxyl-terminal CAAX (C, cysteine; A, aliphatic amino acid; X, any amino acid) motif in which the "X" residue is leucine. The enzyme is composed of a 48-kilodalton alpha subunit and a 43-kilodalton beta subunit. Peptides isolated from the alpha subunit of GGTase-I were shown to be identical with the alpha subunit of a related enzyme, protein farnesyltransferase. Overlapping cDNA clones containing the complete coding sequence for the beta subunit of GGTase-I were obtained from rat and human cDNA libraries. The cDNA clones from both species each predicted a protein of 377 amino acids with molecular masses of 42.4 kilodaltons (human) and 42.5 kilodaltons (rat). Amino acid sequence comparison suggests that the protein encoded by the Saccharomyces cerevisiae gene CDC43 is the yeast counterpart of the mammalian GGTase-I beta subunit. Co-expression of the GGTase-I beta subunit cDNA together with the alpha subunit of protein farnesyltransferase in Escherichia coli produced recombinant GGTase-I with electrophoretic and enzymatic properties indistinguishable from native GGTase-I.
- Published
- 1994
- Full Text
- View/download PDF
39. Isoprenoid diphosphate utilization by recombinant human farnesyl:protein transferase: Interactive binding between substrates and a preferred kinetic pathway
- Author
-
Scott D. Mosser, Michael D. Schaber, Charles A. Omer, Jackson B. Gibbs, Jules A. Shafer, and David L. Pompliano
- Subjects
Alkyl and Aryl Transferases ,biology ,Farnesyl Protein Transferase ,Stereochemistry ,Chemistry ,Farnesyltransferase ,Substrate (chemistry) ,Tritium ,Biochemistry ,Recombinant Proteins ,Substrate Specificity ,Kinetics ,Geranylgeranylation ,Polyisoprenyl Phosphates ,Prenylation ,Transferases ,Escherichia coli ,biology.protein ,Humans ,Transferase ,Computer Simulation ,Enzyme kinetics ,Ternary complex - Abstract
The catalytic utilization of dimethylallyl, geranyl, farnesyl, and geranylgeranyl diphosphates in the reaction catalyzed by recombinant human farnesyl:protein transferase (hFPTase) has been examined in the presence of three different protein substrates, Ras-CVLS, Ras-CVIM, and Ras-CAIL. hFPTase catalyzed both farnesylation and geranylation of Ras-CVLS and of Ras-CVIM but not of Ras-CAIL. Geranylgeranylation was observed, but only when Ras-CVIM was the acceptor substrate. Steady-state initial velocity and dead-end inhibitor studies indicate that hFPTase-catalyzed geranylation, like bovine FPTase-catalyzed farnesylation, proceeds through a random order, sequential mechanism. Surprisingly, however, Michaelis constants for a given protein acceptor substrate varied depending upon which isoprenoid diphosphate was used as the donor substrate, showing that these substrates do not bind independently to the enzyme (under catalytic conditions). In addition, at very high concentrations of Ras-CVIM, substrate inhibition was observed in the presence of both FPP and GPP. Isotope partitioning studies showed that, at high concentrations of Ras-CVIM, more than 80% of the bound farnesyl diphosphate (FPP) can be trapped as product, suggesting that the binary complex is catalytically competent and that the ternary complex proceeds to product faster than it releases FPP. The release rate of FPP from the binary complex was calculated to be 0.05 s-1, which is only about eight times greater than kcat. Thus, the binding of FPP to the enzyme in the presence of the protein substrate is not an equilibrium situation.(ABSTRACT TRUNCATED AT 250 WORDS)
- Published
- 1993
- Full Text
- View/download PDF
40. Isolation and structure of chaetomellic acids A and B from Chaetomella acutiseta: farnesyl pyrophosphate mimic inhibitors of ras farnesyl-protein transferase
- Author
-
Ralph T. Mosley, Rosalind G. Jenkins, Jackson B. Gibbs, Suresh B. Singh, Gerald F. Bills, Mary Nallin-Omstead, Keith C. Silverman, Deborah L. Zink, Georg Albers-Schönberg, Russell B. Lingham, Michael A. Goetz, and Jerrold M. Liesch
- Subjects
chemistry.chemical_classification ,Farnesyl Protein Transferase ,biology ,Stereochemistry ,Farnesyltransferase ,Organic Chemistry ,Farnesyl pyrophosphate ,Biological activity ,Biochemistry ,chemistry.chemical_compound ,Enzyme ,Dicarboxylic acid ,chemistry ,Enzyme inhibitor ,Drug Discovery ,biology.protein ,Transferase - Abstract
Farnesyl-Protein transferase catalyses a post-translational modification of Ras that is obligatory for the cell transforming activity of this oncogene protein. The screening of natural products to identify inhibitors of this enzyme as a potential anticancer agents, has led to the isolation of two novel dicarboxylic acids, named chaetomellic acids from Chaetomella acutiseta, as potent and selective inhibitors which appear to be the first examples of nonphosphorous containing FPP mimics.
- Published
- 1993
- Full Text
- View/download PDF
41. Selective Inhibition of ras -Dependent Transformation by a Farnesyltransferase Inhibitor
- Author
-
Scott D. Mosser, Samuel L. Graham, Jackson B. Gibbs, Edward M. Scolnick, S J deSolms, David L. Pompliano, E A Giuliani, Oliff Allen I, Robert L. Smith, and Nancy E. Kohl
- Subjects
Farnesyl Protein Transferase ,Farnesyltransferase ,Protein Prenylation ,Antineoplastic Agents ,Biology ,Cell Line ,Prenylation ,Transferases ,Animals ,Farnesyltranstransferase ,Oncogene Proteins ,Farnesyl-diphosphate farnesyltransferase ,Alkyl and Aryl Transferases ,Multidisciplinary ,Tetrapeptide ,Farnesyl Transferase Inhibitor ,Farnesyltransferase inhibitor ,Dipeptides ,Rats ,Cell Transformation, Neoplastic ,Genes, ras ,Biochemistry ,Drug Design ,biology.protein ,Cell Division - Abstract
To acquire transforming potential, the precursor of the Ras oncoprotein must undergo farnesylation of the cysteine residue located in a carboxyl-terminal tetrapeptide. Inhibitors of the enzyme that catalyzes this modification, farnesyl protein transferase (FPTase), have therefore been suggested as anticancer agents for tumors in which Ras contributes to transformation. The tetrapeptide analog L-731,735 is a potent and selective inhibitor of FPTase in vitro. A prodrug of this compound, L-731,734, inhibited Ras processing in cells transformed with v-ras. L-731,734 decreased the ability of v-ras-transformed cells to form colonies in soft agar but had no effect on the efficiency of colony formation of cells transformed by either the v-raf or v-mos oncogenes. The results demonstrate selective inhibition of ras-dependent cell transformation with a synthetic organic inhibitor of FPTase.
- Published
- 1993
- Full Text
- View/download PDF
42. Characterization of recombinant human farnesyl-protein transferase: Cloning, expression, farnesyl diphosphate binding, and functional homology with yeast prenyl-protein transferases
- Author
-
Nancy E. Kohl, Jackson B. Gibbs, Ronald E. Diehl, Scott Powers, Charles M. Allen, Astrid M. Kral, Charles A. Omer, and George C. Prendergast
- Subjects
Farnesyl Protein Transferase ,Farnesyltransferase ,Protein subunit ,Molecular Sequence Data ,Saccharomyces cerevisiae ,Gene Expression ,Biology ,Molecular cloning ,Biochemistry ,Structure-Activity Relationship ,Polyisoprenyl Phosphates ,Prenylation ,Isoprenoid binding ,Transferases ,Escherichia coli ,Animals ,Farnesyltranstransferase ,Humans ,Transferase ,Amino Acid Sequence ,Cloning, Molecular ,Alkyl and Aryl Transferases ,Base Sequence ,Sequence Homology, Amino Acid ,DNA ,biology.organism_classification ,Molecular biology ,Recombinant Proteins ,Rats ,Kinetics ,Mutation ,biology.protein ,Cattle ,Sesquiterpenes - Abstract
We have isolated cDNAs encoding the alpha and beta subunits of human farnesyl-protein transferase (FPTase). The proteins encoded by these two cDNAs are 93-95% identical to the corresponding subunits of bovine and rat FPTase and show regions of homology with proteins encoded by Saccharomyces cerevisiae prenyl-protein transferase genes. Human FPTase expressed in Escherichia coli from a translationally coupled operon had kinetic properties similar to those of FPTase isolated from bovine brain. Examination of farnesyl diphosphate binding indicated that while neither individual subunit was capable of isoprenoid binding, a radiolabeled farnesyl diphosphate analog could be specifically photo-cross-linked to the beta subunit of FPTase holoenzyme. To further analyze subunit structure-function and to detect functional similarities with yeast prenyl-protein transferases (FPTase and two geranylgeranyl-protein transferases), amino acid changes homologous to those found in mutant yeast prenyl-protein transferase subunits were made in the subunits of human FPTase. Substitutions in either the alpha or beta subunits that decrease the activity of yeast prenyl-protein transferases were also observed to impair human FPTase. Kinetic analyses showed that these mutant human FPTases have Km and kcat values that are altered with respect to wild-type human FPTase.
- Published
- 1993
- Full Text
- View/download PDF
43. Selective inhibition of farnesyl-protein transferase blocks ras processing in vivo
- Author
-
Scott D. Mosser, Sheo B. Singh, Russell B. Lingham, Jackson B. Gibbs, Rands E, Edward M. Scolnick, David L. Pompliano, Oliff Allen I, and Nancy E. Kohl
- Subjects
chemistry.chemical_classification ,biology ,Farnesyl Protein Transferase ,Farnesyltransferase ,Biological activity ,Zaragozic acid ,Cell Biology ,Biochemistry ,chemistry.chemical_compound ,Enzyme ,chemistry ,Prenylation ,Enzyme inhibitor ,biology.protein ,Transferase ,Molecular Biology - Abstract
The ras oncogene product, Ras, is synthesized in vivo as a precursor protein that requires post-translational processing to become biologically active and to be capable of transforming mammalian cells. Farnesylation appears to be a critical modification of Ras, and thus inhibitors of the farnesyl-protein transferase (FPTase) that catalyzes this reaction may block ras-dependent tumorigenesis. Three structural classes of FPTase inhibitors were identified: (alpha-hydroxyfarnesyl)phosphonic acid, chaetomellic acids, and zaragozic acids. By comparison, these compounds were weaker inhibitors of geranylgeranyl-protein transferases. Each of these inhibitors was competitive with respect to farnesyl diphosphate in the FPTase reaction. All compounds were assayed for inhibition of Ras processing in Ha-ras-transformed NIH3T3 fibroblasts. Ras processing was inhibited by 1 microM (alpha-hydroxyfarnesyl)phosphonic acid. Neither chaetomellic acid nor zaragozic acid were active in this assay. These results are the first demonstration that a small organic chemical selected for inhibition of FPTase can inhibit Ras processing in vivo.
- Published
- 1993
- Full Text
- View/download PDF
44. ChemInform Abstract: Isolation and Structure of Chaetomellic Acids A and B from Chaetomella acutiseta: Farnesyl Pyrophosphate Mimic Inhibitors of Ras Farnesyl- Protein Transferase
- Author
-
Georg Albers-Schönberg, Suresh B. Singh, Keith C. Silverman, Michael A. Goetz, Russell B. Lingham, Jerrold M. Liesch, Gerald F. Bills, Ralph T. Mosley, Rosalind G. Jenkins, Jackson B. Gibbs, Mary Nallin-Omstead, and Deborah L. Zink
- Subjects
chemistry.chemical_classification ,Farnesyl Protein Transferase ,Oncogene ,Cell ,Farnesyl pyrophosphate ,General Medicine ,Chaetomella acutiseta ,chemistry.chemical_compound ,Enzyme ,medicine.anatomical_structure ,chemistry ,Biochemistry ,medicine ,Transferase - Abstract
Farnesyl-Protein transferase catalyses a post-translational modification of Ras that is obligatory for the cell transforming activity of this oncogene protein. The screening of natural products to identify inhibitors of this enzyme as a potential anticancer agents, has led to the isolation of two novel dicarboxylic acids, named chaetomellic acids from Chaetomella acutiseta, as potent and selective inhibitors which appear to be the first examples of nonphosphorous containing FPP mimics.
- Published
- 2010
- Full Text
- View/download PDF
45. ChemInform Abstract: Fusidienol (I): A Novel Inhibitor of Ras Farnesyl-Protein Transferase from Fusidium griseum
- Author
-
Michael A. Goetz, Rosalind G. Jenkins, E. T. T. Jones, Jackson B. Gibbs, Mary Nallin-Omstead, Keith C. Silverman, Sheo B. Singh, Russell B. Lingham, and Gerald F. Bills
- Subjects
Biochemistry ,Farnesyl Protein Transferase ,Chemistry ,General Medicine ,Fusidienol ,Fusidium griseum - Published
- 2010
- Full Text
- View/download PDF
46. ChemInform Abstract: Generation and Screening of Solution-Phase Synthetic Peptide Combinatorial Libraries
- Author
-
Andrew Wallace, Kenneth S. Koblan, Riccardo Cortese, Jackson B. Gibbs, and Antonello Pessi
- Subjects
General Medicine - Published
- 2010
- Full Text
- View/download PDF
47. ChemInform Abstract: Imidazole-Containing Diarylether and Diarylsulfone Inhibitors of Farnesyl-Protein Transferase
- Author
-
Robert B. Lobell, J. Christopher Culberson, Jackson B. Gibbs, Christopher J. Dinsmore, Kenneth S. Koblan, Theresa M. Williams, Nancy E. Kohl, Dongming Liu, Rands E, and Timothy J. O'Neill
- Subjects
chemistry.chemical_compound ,Farnesyl Protein Transferase ,chemistry ,Stereochemistry ,Imidazole ,General Medicine - Published
- 2010
- Full Text
- View/download PDF
48. Steady-state kinetic mechanism of ras farnesyl:protein transferase
- Author
-
Michael D. Schaber, Neville J. Anthony, David L. Pompliano, Elaine Rands, Scott D. Mosser, and Jackson B. Gibbs
- Subjects
Farnesyl Protein Transferase ,Stereochemistry ,Farnesyltransferase ,Biochemistry ,Catalysis ,Substrate Specificity ,Non-competitive inhibition ,Polyisoprenyl Phosphates ,Transferases ,Animals ,Enzyme kinetics ,Ternary complex ,Alkyl and Aryl Transferases ,biology ,Chemistry ,Brain ,Substrate (chemistry) ,Kinetics ,Product inhibition ,biology.protein ,Cattle ,Electrophoresis, Polyacrylamide Gel ,Chromatography, Thin Layer ,Steady state (chemistry) ,Oligopeptides ,Sesquiterpenes - Abstract
The steady-state kinetic mechanism of bovine brain farnesyl:protein transferase (FPTase) has been determined using a series of initial velocity studies, including both dead-end substrate and product inhibitor experiments. Reciprocal plots of the initial velocity data intersected on the 1/[s] axis, indicating that a ternary complex forms (sequential mechanism) and suggesting that the binding of one substrate does not affect the binding of the other. The order of substrate addition was probed by determining the patterns of dead-end substrate and product inhibition. Two nonhydrolyzable analogues of farnesyl diphosphate, (alpha-hydroxyfarnesyl)phosphonic acid (1) and [[(farnesylmethyl)hydroxyphosphinyl]methyl]phosphonic acid (2), were both shown to be competitive inhibitors of farnesyl diphosphate and noncompetitive inhibitors of Ras-CVLS. Four nonsubstrate tetrapeptides, CV[D-L]S, CVLS-NH2, N-acetyl-L-penicillamine-VIM, and CIFM, were all shown to be noncompetitive inhibitors of farnesyl diphosphate and competitive inhibitors of Ras-CVLS. These data are consistent with random order of substrate addition. Product inhibition patterns corroborated the results found with the dead-end substrate inhibitors. We conclude that bovine brain FPTase proceeds through a random order sequential mechanism. Determination of steady-state parameters for several physiological Ras-CaaX variants showed that amino acid changes affected the values of KM, but not those of kcat, suggesting that the catalytic efficiencies (kcat/KM) of Ras-CaaX substrates depend largely upon their relative binding affinity for FPTase.
- Published
- 1992
- Full Text
- View/download PDF
49. Role of GTPase activating protein in mitogenic signalling through phosphatidylcholine-hydrolysing phospholipase C
- Author
-
F. McCormick, Isabel Dominguez, Maria T. Diaz-Meco, G. Graziani, M E Cornet, Jackson B. Gibbs, Jorge Moscat, Terje Johansen, M.S. Marshall, and A Garcia de Herreros
- Subjects
GTPase-activating protein ,Maturation-Promoting Factor ,Maturation promoting factor ,Protamine Kinase ,GTPase ,Oncogene Protein p21(ras) ,General Biochemistry, Genetics and Molecular Biology ,Xenopus laevis ,Bacillus cereus ,Anti-apoptotic Ras signalling cascade ,CDC2 Protein Kinase ,Animals ,Phosphorylation ,Molecular Biology ,General Immunology and Microbiology ,Phospholipase C ,biology ,Effector ,Hydrolysis ,General Neuroscience ,GTPase-Activating Proteins ,Proteins ,Recombinant Proteins ,Biochemistry ,ras GTPase-Activating Proteins ,Type C Phospholipases ,Phosphatidylcholines ,biology.protein ,Mitogens ,Signal transduction ,Signal Transduction ,Research Article - Abstract
Recent evidence has accumulated showing that activation of PLC-catalysed hydrolysis of phosphatidylcholine (PC-PLC) is a critical step in mitogenic signal transduction both in fibroblasts and in oocytes from Xenopus laevis. The products of ras genes activate PC-PLC, bind guanine nucleotides, have intrinsic GTPase activity, and are regulated by a GTPase-activating protein (GAP). It has been suggested that, in addition to its regulatory properties, GAP may also be necessary for ras function as a downstream effector molecule. In this study, evidence is presented that strongly suggests that the functional interaction between ras p21 and GAP is sufficient and necessary for activation of maturation promoting factor (MPF) H1-kinase activity in oocytes, and that PC hydrolysis is critically involved in this mechanism. Therefore, we identify GAP as a further step required for signalling through PC-PLC, and necessary for the control of oocyte maturation in response to ras p21/insulin but not to progesterone.
- Published
- 1991
- Full Text
- View/download PDF
50. Sequence dependence of protein isoprenylation
- Author
-
Elaine Rands, Michael D. Schaber, Scott D. Mosser, David L. Pompliano, Mark S. Marshall, Victor M. Garsky, S L Moores, M. B. O'hara, and Jackson B. Gibbs
- Subjects
chemistry.chemical_classification ,Geranylgeranyl Transferase ,Fungal protein ,Farnesyl Protein Transferase ,Protein subunit ,Farnesyltransferase ,C-terminus ,Cell Biology ,Biology ,Biochemistry ,Amino acid ,chemistry ,biology.protein ,Molecular Biology ,Peptide sequence - Abstract
Several proteins have been shown to be post-translationally modified on a specific C-terminal cysteine residue by either of two isoprenoid biosynthetic pathway metabolites, farnesyl diphosphate or geranylgeranyl diphosphate. Three enzymes responsible for protein isoprenylation were resolved chromatographically from the cytosolic fraction of bovine brain: a farnesyl-protein transferase (FTase), which modified the cell-transforming Ras protein, and two geranyl-geranyl-protein transferases, one (GGTase-I) which modified a chimeric Ras having the C-terminal amino acid sequence of the gamma-6 subunit of heterotrimeric GTP-binding proteins, and the other (GGTase-II) which modified the Saccharomyces cerevisiae secretory GTPase protein YPT1. In a S. cerevisiae strain lacking FTase activity (ram1), both GGTases were detected at wild-type levels. In a ram2 S. cerevisiae strain devoid of FTase activity, GGTase-I activity was reduced by 67%, suggesting that GGTase-I and FTase activities derive from different enzymes but may share a common genetic feature. For the FTase and the GGTase-I activities, the C-terminal amino acid sequence of the protein substrate, the CAAX box, appeared to contain all the critical determinants for interaction with the transferase. In fact, tetrapeptides with amino acid sequences identical to the C-terminal sequences of the protein substrates for FTase or GGTase-I competed for protein isoprenylation by acting as alternative substrates. Changes in the CAAX amino acid sequence of protein substrates markedly altered their ability to serve as substrates for both FTase and GGTase-I. In addition, it appeared that FTase and GGTase-I had complementary affinities for CAAX protein substrates; that is, CAAX proteins that were good substrates for FTase were, in general, poor substrates for GGTase-I, and vice versa. In particular, a leucine residue at the C terminus influenced whether a CAAX protein was either farnesylated or geranylgeranylated preferentially. The YPT1 C terminus peptide, TGGGCC, did not compete or serve as a substrate for GGTase-II, indicating that the interaction between GGTase-II and YPT1 appeared to depend on more than the 6 C-terminal residues of the protein substrate sequence. These results identify three different isoprenyl-protein transferases that are each selective for their isoprenoid and protein substrates.
- Published
- 1991
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.