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










Publication year range
1.
Antibiotics (Basel) ; 7(4)2018 Oct 26.
Article in English | MEDLINE | ID: mdl-30373130

ABSTRACT

The use of silver to control infections was common in ancient civilizations. In recent years, this material has resurfaced as a therapeutic option due to the increasing prevalence of bacterial resistance to antimicrobials. This renewed interest has prompted researchers to investigate how the antimicrobial properties of silver might be enhanced, thus broadening the possibilities for antimicrobial applications. This review presents a compilation of patented products utilizing any forms of silver for its bactericidal actions in the decade 2007⁻2017. It analyses the trends in patent applications related to different forms of silver and their use for antimicrobial purposes. Based on the retrospective view of registered patents, statements of prognosis are also presented with a view to heightening awareness of potential industrial and health care applications.

2.
Eur J Med Chem ; 120: 353-62, 2016 Sep 14.
Article in English | MEDLINE | ID: mdl-27236016

ABSTRACT

Diarrhoeal diseases caused by the intestinal parasites Giardia lamblia and Entamoeba histolytica constitute a major global health burden. Nitroimidazoles are first-line drugs for the treatment of giardiasis and amebiasis, with metronidazole 1 being the most commonly used drug worldwide. However, treatment failures in giardiasis occur in up to 20% of cases and development of resistance to metronidazole is of concern. We have re-examined 'old' nitroimidazoles as a foundation for the systematic development of next-generation derivatives. Using this approach, derivatisation of the nitroimidazole carboxamide scaffold provided improved antiparasitic agents. Thirty-three novel nitroimidazole carboxamides were synthesised and evaluated for activity against G. lamblia and E. histolytica. Several of the new compounds exhibited potent activity against G. lamblia strains, including metronidazole-resistant strains of G. lamblia (EC50 = 0.1-2.5 µM cf. metronidazole EC50 = 6.1-18 µM). Other compounds showed improved activity against E. histolytica (EC50 = 1.7-5.1 µM cf. metronidazole EC50 = 5.0 µM), potent activity against Trichomonas vaginalis (EC50 = 0.6-1.4 µM cf. metronidazole EC50 = 0.8 µM) and moderate activity against the intestinal bacterial pathogen Clostridium difficile (0.5-2 µg/mL, cf. metronidazole = 0.5 µg/mL). The new compounds had low toxicity against mammalian kidney and liver cells (CC50 > 100 µM), and selected antiparasitic hits were assessed for human plasma protein binding and metabolic stability in liver microsomes to demonstrate their therapeutic potential.


Subject(s)
Antiparasitic Agents/pharmacology , Nitroimidazoles/pharmacology , Parasites/drug effects , Animals , Cells, Cultured , Drug Resistance , Drug Stability , Entamoeba histolytica/drug effects , Giardia lamblia/drug effects , Humans , Trichomonas vaginalis/drug effects
3.
Br J Cancer ; 111(5): 894-902, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-24983364

ABSTRACT

BACKGROUND: The binding of STAT3 and STAT5 to growth factor and cytokine receptors such as EGFR and IL-6 receptor gp130 is critical to their activation and ability to contribute to malignant transformation. Therefore, interfering with these biochemical processes could lead to the discovery of novel anticancer agents. METHODS: Co-immunoprecipitation, western blotting, microscopy, DNA binding, invasion, and soft agar assays as well as a mouse model were used to investigate the mechanism by which the natural product Withacnistin (Wit) inhibits STAT 3/5 tyrosine phosphoryaltion and activation. RESULTS: Wit blocks EGF- and IL-6-stimulated binding of STAT3 and STAT5 to EGFR and gp130. Wit inhibits EGF-, PDGF-, IL-6-, IFNß-, and GM-CSF-stimulation of tyrosine phosphorylation of STAT3 and STAT5 but not of EGFR or PDGFR. The inhibition of P-STAT3 and P-STAT5 occurred rapidly, within minutes of Wit treatment and growth factor stimulation. Wit also inhibits STAT3 nuclear translocation, DNA binding, promoter transcriptional activation, and it suppresses the expression levels of STAT3 target genes such as Bcl-xL and Mcl-1. Finally, Wit induces apoptosis, inhibits anchorage-dependent and -independent growth and invasion, and causes breast tumour regression in an ErbB2-driven transgenic mouse model. CONCLUSIONS: These data warrant further development of Wit as a novel anticancer drug for targeting tumours that harbour hyperactivated STAT3 and STAT5.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Ergosterol/analogs & derivatives , Intercellular Signaling Peptides and Proteins/metabolism , Lactones/pharmacology , Receptors, Cytokine/metabolism , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cytokine Receptor gp130/metabolism , DNA-Binding Proteins , Epidermal Growth Factor/metabolism , Ergosterol/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interferon-beta/metabolism , Interleukin-6/metabolism , Mice , Mice, Transgenic , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , NIH 3T3 Cells , Phosphorylation/drug effects , Platelet-Derived Growth Factor/metabolism , bcl-X Protein/metabolism
4.
Oncogene ; 26(5): 633-40, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-16909123

ABSTRACT

Recently, we have shown that RhoB suppresses EGFR-, ErbB2-, Ras- and Akt-mediated malignant transformation and metastasis. In this paper, we demonstrate that the novel antitumor agents farnesyltransferase inhibitors (FTIs) and geranylgeranyltransferase I inhibitors (GGTIs) upregulate RhoB expression in a wide spectrum of human cancer cells including those from pancreatic, breast, lung, colon, bladder and brain cancers. RhoB induction by FTI-277 and GGTI-298 occurs at the transcriptional level and is blocked by actinomycin D. Reverse transcription-PCR experiments documented that the increase in RhoB protein levels is due to an increase in RhoB transcription. Furthermore, treatment with FTIs and GGTIs of cancer cells results in HDAC1 dissociation, HAT association and histone acetylation of the RhoB promoter. Thus, promoter acetylation is a novel mechanism by which RhoB expression levels are regulated following treatment with the anticancer agents FTIs and GGTIs.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Farnesyltranstransferase/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Histone Deacetylases/metabolism , Histones/metabolism , Promoter Regions, Genetic , rhoB GTP-Binding Protein/genetics , Acetylation , Alkyl and Aryl Transferases/metabolism , Antineoplastic Agents , Benzamides/pharmacology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/metabolism , Histone Deacetylase 1 , Humans , Methionine/analogs & derivatives , Methionine/pharmacology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Protein Processing, Post-Translational , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Up-Regulation , rhoB GTP-Binding Protein/metabolism
5.
Bioorg Med Chem Lett ; 11(6): 761-4, 2001 Mar 26.
Article in English | MEDLINE | ID: mdl-11277514

ABSTRACT

Malaria continues to represent a very serious health problem in the tropics. The current methods of clinical treatment are showing deficiencies due to the increased incidence of resistance in the parasite. In the present paper we report the design, synthesis, and evaluation of potential antimalarial agents against a novel target, protein farnesyltransferase. We show that the most potent compounds are active against Plasmodium falciparum in vitro at submicromolar concentrations.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antimalarials/pharmacology , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Plasmodium falciparum/drug effects , Alkyl and Aryl Transferases/metabolism , Animals , Antimalarials/chemical synthesis , Antimalarials/chemistry , Drug Design , Drug Resistance , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Imidazoles/chemical synthesis , Imidazoles/chemistry , Inhibitory Concentration 50 , Parasitic Sensitivity Tests , Structure-Activity Relationship
6.
Nat Biotechnol ; 18(10): 1065-70, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11017044

ABSTRACT

We have designed a molecule, GFB-111, that binds to platelet-derived growth factor (PDGF), prevents it from binding to its receptor tyrosine kinase, and blocks PDGF-induced receptor autophosphorylation, activation of Erk1 and Erk2 kinases, and DNA synthesis. GFB-111 is highly potent (IC50 = 250 nM) and selective for PDGF over EGF, IGF-1, aFGF, bFGF, and HRGbeta (IC50 values > 100 microM), but inhibits VEGF-induced Flk-1 tyrosine phosphorylation and Erk1/Erk2 activation with an IC50 of 10 microM. GFB-111 treatment of nude mice bearing human tumors resulted in significant inhibition of tumor growth and angiogenesis. The results demonstrate the feasibility of designing novel growth factor-binding molecules with potent anticancer and antiangiogenic activity.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Drug Design , Glioblastoma/drug therapy , Peptides, Cyclic/pharmacology , Platelet-Derived Growth Factor/antagonists & inhibitors , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Cell Line , DNA/biosynthesis , Endothelial Growth Factors/antagonists & inhibitors , Endothelial Growth Factors/pharmacology , Enzyme Activation/drug effects , Glioblastoma/blood supply , Glioblastoma/pathology , Humans , Inhibitory Concentration 50 , Lymphokines/antagonists & inhibitors , Lymphokines/pharmacology , Mice , Mice, Nude , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Transplantation , Neovascularization, Pathologic/drug therapy , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , Peptides, Cyclic/therapeutic use , Phosphorylation/drug effects , Platelet-Derived Growth Factor/metabolism , Platelet-Derived Growth Factor/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Substrate Specificity , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
7.
Am J Physiol Lung Cell Mol Physiol ; 278(2): L329-34, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10666117

ABSTRACT

We recently showed that the farnesyltransferase inhibitor FTI-277 blocks interleukin 1beta (IL-1beta)-induced nitric oxide production in pulmonary vascular smooth muscle cells (SMC), whereas the geranylgeranyltransferase inhibitor GGTI-298 enhances this effect. Here we show that IL-1beta and platelet-derived growth factor (PDGF) stimulate superoxide production by pulmonary vascular SMC and that this effect is blocked by both FTI-277 and GGTI-298, suggesting that farnesylated and geranylgeranylated proteins are required for superoxide production. We also show that FTI-277 and GGTI-298 block superoxide production stimulated by constitutively active mutant H-Ras. Furthermore, superoxide production by IL-1beta, PDGF factor, and constitutively activated Ras is blocked by diphenyleneiodonium, implicating NAD(P)H oxidase as the generating enzyme. Given the role of oxidant radicals in vascular reactivity and injury, the action of both FTI-277 and GGTI-298 in suppressing superoxide generation by an inflammatory cytokine as well as by a potent smooth muscle mitogen may be therapeutically useful.


Subject(s)
Dimethylallyltranstransferase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Muscle, Smooth, Vascular/metabolism , Pulmonary Circulation/physiology , Superoxides/antagonists & inhibitors , Animals , Benzamides/pharmacology , Cells, Cultured , Genes, ras/genetics , Interleukin-1/pharmacology , Male , Methionine/analogs & derivatives , Methionine/pharmacology , Muscle, Smooth, Vascular/cytology , Mutation/physiology , Platelet-Derived Growth Factor/pharmacology , Rats , Superoxides/metabolism , Transfection
8.
Cancer Res ; 59(19): 4919-26, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10519405

ABSTRACT

Ras malignant transformation requires posttranslational modification by farnesyltransferase (FTase). Here we report on the design and antitumor activity, in monotherapy as well as in combination therapy with cytotoxic agents, of a novel class of non-thiol-containing peptidomimetic inhibitors of FTase and the closely related family member geranylgeranyltransferase I (GGTase I). The non-thiol-containing FTI-2148 is highly selective for FTase (IC50, 1.4 nM) over GGTase I (IC50, 1700 nM), whereas GGTI-2154 is highly selective for GGTase I (21 nM) over FTase (IC50, 5600 nM). In whole cells, the corresponding methylester prodrug FTI-2153 is >3000-fold more potent at inhibiting H-Ras (IC50, 10 nM) than Rap1A processing, whereas GGTI-2166 is over 100-fold more selective at inhibiting Rap1A (IC50, 300 nM) over H-Ras processing. Furthermore, FTI-2153 was highly effective at suppressing oncogenic H-Ras constitutive activation of mitogen-activated protein kinase and human tumor growth in soft agar. FTI-2148 suppressed the growth of the human lung adenocarcinoma A-549 cells in nude mice by 33, 67, and 91% in a dose-dependent manner. Combination therapy of FTI-2148 with either cisplatin, gemcitabine, or Taxol resulted in a greater antitumor efficacy than monotherapy. GGTI-2154 in similar antitumor efficacy experiments is less potent than FTI-2148 and inhibits tumor growth by 9, 27, and 46%. Combination therapy of GGTI-2154 with cisplatin, gemcitabine, or Taxol is also more effective. Finally, FTI-2148 and GGTI-2154 are 30- and 33-fold more selective and 30- and 16-fold more potent in whole cells than our previously reported thiol-containing FTI-276 and GGTI-297, respectively. Thus, our results demonstrate that this highly potent and selective novel class of non-thiol-containing peptidomimetics inhibits human tumor growth in whole animals and that combination therapy with cytotoxic agents is more beneficial than monotherapy.


Subject(s)
Adenocarcinoma/drug therapy , Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzamides/chemistry , Benzamides/therapeutic use , Enzyme Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Oligopeptides/chemistry , 3T3 Cells , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Antineoplastic Combined Chemotherapy Protocols/toxicity , Benzamides/toxicity , Cell Division/drug effects , Cisplatin/administration & dosage , Cisplatin/therapeutic use , Cisplatin/toxicity , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Deoxycytidine/toxicity , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/toxicity , Farnesyltranstransferase , Humans , Mice , Mice, Nude , Mitogen-Activated Protein Kinases/drug effects , Molecular Structure , Paclitaxel/administration & dosage , Paclitaxel/therapeutic use , Paclitaxel/toxicity , Transplantation, Heterologous , Tumor Cells, Cultured , rap1 GTP-Binding Proteins/antagonists & inhibitors , ras Proteins/antagonists & inhibitors , Gemcitabine
9.
J Med Chem ; 42(8): 1333-40, 1999 Apr 22.
Article in English | MEDLINE | ID: mdl-10212118

ABSTRACT

The design, synthesis, and biological evaluation of a family of peptidomimetic inhibitors of protein geranylgeranyltransferase-I (PGGTase-I) are reported. The inhibitors are based on the C-terminal CAAL sequence of many geranylgeranylated proteins. Using 2-aryl-4-aminobenzoic acid derivatives as mimetics for the central dipeptide (AA), we have attached a series of imidazole and pyridine derivatives to the N-terminus as cysteine replacements. These non-thiol-containing peptidomimetics show exceptional selectivity for PGGTase-I over the closely related enzyme protein farnesyltransferase (PFTase). This selectivity is retained in whole cells where the inhibitors were shown to block the geranylgeranylation of Rap-1A without affecting the farnesylation of small GTP-binding proteins such as Ras.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Imidazoles/chemical synthesis , Leucine/analogs & derivatives , Leucine/chemical synthesis , Protein Prenylation/drug effects , 3T3 Cells , Animals , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , GTP-Binding Proteins/biosynthesis , Imidazoles/chemistry , Imidazoles/pharmacology , Leucine/chemistry , Leucine/pharmacology , Mice , Molecular Mimicry , Structure-Activity Relationship , rap GTP-Binding Proteins
10.
Biochem Biophys Res Commun ; 252(1): 111-6, 1998 Nov 09.
Article in English | MEDLINE | ID: mdl-9813154

ABSTRACT

The TGF-betas are multipotent in their biological activity, modulating cell growth and differentiation as well as extracellular matrix deposition and degradation. Most of these activities involve modulation of gene transcription. However, TGF-beta1 has been shown previously to substantially increase the expression of elastin by stabilization of tropoelastin mRNA through a signaling pathway which involves a phosphatidylcholine-specific phospholipase and a protein kinase C. The present results, through the use of specific inhibitors of geranylgeranyl transferase I, farnesyl transferase, and acyl transferase, demonstrate that geranylgeranylated and acylated, but not farnesyslated protein(s) is required for this TGF-beta1 effect. In addition, the general tyrosine kinase inhibitor genistein completely blocked this TGF-beta1 effect. The results suggest that the TGF-beta1 signaling pathway requires not only receptor ser/thr kinase activity, but also tyrosine kinase and small GTPase activities.


Subject(s)
Acyltransferases/metabolism , Alkyl and Aryl Transferases/metabolism , Elastin/genetics , RNA, Messenger/metabolism , Signal Transduction/physiology , Transcription, Genetic , Transforming Growth Factor beta/pharmacology , Tropoelastin/genetics , Benzamides/pharmacology , Cell Line , Cholera Toxin/pharmacology , Enzyme Inhibitors/pharmacology , Fibroblasts , GTP Phosphohydrolases/metabolism , Humans , Lung , Methionine/analogs & derivatives , Methionine/pharmacology , Protein Kinase C/metabolism , Protein Prenylation/drug effects , Protein Prenylation/physiology , Transcription, Genetic/drug effects , Type C Phospholipases/metabolism , Virulence Factors, Bordetella/pharmacology
11.
Am J Physiol ; 275(1): L55-63, 1998 07.
Article in English | MEDLINE | ID: mdl-9688935

ABSTRACT

The activity of small GTP-binding proteins is regulated by a critical step in posttranslational processing, namely, the addition of isoprenoid lipids farnesyl and geranylgeranyl, mediated by the enzymes farnesyltransferase (FTase) and geranylgeranyltransferase I (GGTase I), respectively. We have developed compounds that inhibit these enzymes specifically and in this study sought to determine their effects on smooth muscle cells (SMC) from the pulmonary microvasculature. We found that the GGTase I inhibitor GGTI-298 suppressed protein geranylgeranylation and blocked serum-dependent growth as measured by thymidine uptake and cell counts. In the absence of serum, however, GGTI-298 induced apoptosis in these cells as measured by both DNA staining and flow cytometry. The FTase inhibitor FTI-277 selectively inhibited protein farnesylation but had a minor effect on growth and no effect on apoptosis. To further investigate the role of geranylgeranylated proteins in apoptosis, we added the cholesterol synthesis inhibitor lovastatin, which inhibits the biosynthesis of farnesyl and geranylgeranyl pyrophosphates. This also induced apoptosis, but when geranylgeraniol was added to replenish cellular pools of geranylgeranyl pyrophosphate, apoptosis was reduced to baseline. In contrast, farnesol achieved only partial rescue of the cells. These results imply that geranylgeranylated proteins are required for growth and protect SMC against apoptosis. GGTase I inhibitors may be useful in preventing hyperplastic remodeling and may have the potential to induce the apoptotic regression of established vascular lesions.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Apoptosis/physiology , Benzamides/pharmacology , Enzyme Inhibitors/pharmacology , Methionine/analogs & derivatives , Microcirculation/physiology , Muscle, Smooth, Vascular/physiology , Pulmonary Artery/physiology , ras Proteins/biosynthesis , Animals , Apoptosis/drug effects , Cell Division/drug effects , Cells, Cultured , Farnesyltranstransferase , Flow Cytometry , GTP-Binding Proteins/biosynthesis , Kinetics , Lovastatin/pharmacology , Methionine/pharmacology , Microcirculation/cytology , Microcirculation/drug effects , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Protein Prenylation/drug effects , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , Rats , Transcription Factors/biosynthesis , rap GTP-Binding Proteins
12.
Bioorg Med Chem Lett ; 8(17): 2321-6, 1998 Sep 08.
Article in English | MEDLINE | ID: mdl-9873535

ABSTRACT

The general approach of using a bicyclic template to produce inhibitors of the protease superfamily of enzymes has been investigated. The Diels Alder cycloaddition reaction on solid support has been found to be highly efficient for the synthesis of libraries of compounds that mimic the beta-strand secondary structure of proteins. Several potent and selective inhibitors of proteases have been discovered.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis , Peptide Library , Protease Inhibitors/chemical synthesis , Protein Structure, Secondary , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Chymases , Indicators and Reagents , Kallikreins/antagonists & inhibitors , Kinetics , Models, Molecular , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacology , Structure-Activity Relationship , Thrombin/antagonists & inhibitors , Trypsin Inhibitors/chemical synthesis , Trypsin Inhibitors/chemistry , Trypsin Inhibitors/pharmacology , Tryptases
13.
J Biol Chem ; 272(21): 13484-8, 1997 May 23.
Article in English | MEDLINE | ID: mdl-9153192

ABSTRACT

Recently, we have designed farnesyltransferase and geranylgeranyltransferase I inhibitors (FTI-277 and GGTI-298) that selectively block protein farnesylation and geranylgeranylation, respectively. In this study, we describe the opposing effects of these inhibitors on interleukin-1beta (IL-1beta)-stimulated induction of nitric-oxide synthase-2 (NOS-2) in rat pulmonary artery smooth muscle cells (RPASMC) and rat hepatocytes. Pretreatment of cells with GGTI-298 caused a superinduction of NOS-2 by IL-1beta. RPASMC treated with GGTI-298 (10 microM) prior to IL-1beta (10 ng/ml) expressed levels of NOS-2 protein five times higher than those exposed to IL-1beta alone. This superinduction of NOS-2 protein by pretreatment with GGTI-298 resulted in nitrite concentrations in the medium that were 5-fold higher at 10 ng/ml IL-1beta and 10-fold higher at 1 ng/ml IL-1beta. Furthermore, NOS-2 mRNA levels in RPASMC were also increased 6- and 14-fold (at 10 and 1 ng/ml IL-1beta, respectively) when the cells were pretreated with GGTI-298. In contrast, treatment of cells with the inhibitor of protein farnesylation, FTI-277 (10 microM), blocked IL-1beta-induced NOS-2 expression at mRNA and protein levels. Pretreatment with lovastatin, an inhibitor of protein prenylation, resulted in superinduction of NOS-2. This superinduction was reversed by geranylgeraniol, but not by farnesol, further confirming that inhibition of geranylgeranylation, not farnesylation, is responsible for enhanced NOS-2 expression. The results demonstrate that a farnesylated protein(s) mediates IL-1beta induction of NOS-2, whereas a geranylgeranylated protein(s) represses this induction.


Subject(s)
Alkyl and Aryl Transferases , Enzyme Inhibitors/pharmacology , Interleukin-1/pharmacology , Muscle, Smooth, Vascular/enzymology , Nitric Oxide Synthase/biosynthesis , Protein Prenylation , Transferases/antagonists & inhibitors , Animals , Benzamides/pharmacology , Enzyme Induction/drug effects , Farnesyltranstransferase , GTP-Binding Proteins/metabolism , Liver/metabolism , Male , Methionine/analogs & derivatives , Methionine/pharmacology , Muscle, Smooth, Vascular/drug effects , Nitrites/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factors/metabolism , rap GTP-Binding Proteins , ras Proteins/metabolism
14.
Arch Biochem Biophys ; 326(2): 233-7, 1996 Feb 15.
Article in English | MEDLINE | ID: mdl-8611028

ABSTRACT

Lovastatin, a cholesterol biosynthesis inhibitor, has recently been shown to inhibit mitogenesis and tumor growth. We have investigated the effects of lovastatin on the activation of MAP kinase by insulin using as a model HIRcB cells, a rat fibroblast cell line that overexpresses the human insulin receptor. Treatment with lovastatin (1-30 microM) for 24 h decreased the level of activation of MAP kinase by insulin by as much as 60%. Immunoblotting experiments using a specific anti-MAP kinase monoclonal antibody demonstrated that the amount of MAP kinase protein in the cells was not altered by lovastatin treatment. Likewise, lovastatin had no apparent effects on the expression of the insulin receptor. Treatment with lovastatin (20 microM) reduced the percentage of farnesylated Ras by 50%. Immunoprecipitation of tyrosine phosphorylated proteins from HIRcB cell lysates followed by immunodetection of MAP kinase using specific antibodies demonstrated a reduced level of insulin-induced tyrosine phosphorylation levels of MAP kinase in lovastatin-treated cells. Furthermore, immunodetection of the beta-subunit of the insulin receptor in anti-phosphotyrosine immunoprecipitates revealed that treatment with lovastatin reduced the tyrosine phosphorylation levels of the receptor. Lysates obtained from cells treated with increasing concentrations of lovastatin demonstrated a dose-dependent inhibition of the insulin-induced tyrosine phosphorylation of the receptor. Treatment with mevalonic acid prevented the effects of lovastatin demonstrating that the effects of the drug are a consequence of its inhibitory effects on the synthesis of steroids. It is concluded that, in addition to inhibition of Ras farnesylation, lovastatin reduces receptor tyrosine phosphorylation levels which also contributes to the blockade of MAPK activation by the insulin receptor.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Insulin/pharmacology , Lovastatin/pharmacology , Animals , Cell Line , Enzyme Activation/drug effects , Fibroblasts , Humans , Mevalonic Acid/pharmacology , Mitogen-Activated Protein Kinase 1 , Phosphorylation , Protein Processing, Post-Translational/drug effects , Protein-Tyrosine Kinases/metabolism , Rats , Receptor, Insulin/drug effects , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Tyrosine/metabolism , ras Proteins/metabolism
15.
J Biol Chem ; 270(45): 26802-6, 1995 Nov 10.
Article in English | MEDLINE | ID: mdl-7592920

ABSTRACT

Ras-induced malignant transformation requires Ras farnesylation, a lipid posttranslational modification catalyzed by farnesyltransferase (FTase). Inhibitors of this enzyme have been shown to block Ras-dependent transformation, but the mechanism by which this occurs remains largely unknown. We have designed FTI-276, a peptide mimetic of the COOH-terminal Cys-Val-Ile-Met of K-Ras4B that inhibited potently FTase in vitro (IC50 = 500 pM) and was highly selective for FTase over geranylgeranyltransferase I (GGTase I) (IC50 = 50 nM). FTI-277, the methyl ester derivative of FTI-276, was extremely potent (IC50 = 100 nM) at inhibiting H-Ras, but not the geranylgeranylated Rap1A processing in whole cells. Treatment of H-Ras oncogene-transformed NIH 3T3 cells with FTI-277 blocked recruitment to the plasma membrane and subsequent activation of the serine/threonine kinase c-Raf-1 in cells transformed by farnesylated Ras (H-RasF), but not geranylgeranylated, Ras (H-RasGG). FTI-277 induced accumulation of cytoplasmic non-farnesylated H-Ras that was able to bind Raf and form cytoplasmic Ras/Raf complexes in which Raf kinase was not activated. Furthermore, FTI-277 blocked constitutive activation of mitogen-activated protein kinase (MAPK) in H-RasF, but not H-RasGG, or Raf-transformed cells. FTI-277 also inhibited oncogenic K-Ras4B processing and constitutive activation of MAPK, but the concentrations required were 100-fold higher than those needed for H-Ras inhibition. The results demonstrate that FTI-277 blocks Ras oncogenic signaling by accumulating inactive Ras/Raf complexes in the cytoplasm, hence preventing constitutive activation of the MAPK cascade.


Subject(s)
Alkyl and Aryl Transferases , Enzyme Inhibitors/pharmacology , Methionine/analogs & derivatives , Oligopeptides/pharmacology , ras Proteins/metabolism , Amino Acid Sequence , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , Cell Transformation, Neoplastic/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Farnesyltranstransferase , Humans , Methionine/chemical synthesis , Methionine/chemistry , Methionine/pharmacology , Molecular Sequence Data , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-raf , Signal Transduction/drug effects , Transferases/antagonists & inhibitors , ras Proteins/genetics
16.
Biochem Biophys Res Commun ; 214(1): 295-303, 1995 Sep 05.
Article in English | MEDLINE | ID: mdl-7669049

ABSTRACT

Cysteine farnesylation of the Ras carboxyl terminal tetrapeptide CAAX motif (where C = cysteine, A = leucine, isoleucine, or valine, and X = methionine or serine) is required for Ras biological activity. In this report, we describe the effects of inhibitors of farnesyltransferase (FTase), the enzyme responsible for this lipid modification, on platelet-derived growth factor (PDGF) signaling in NIH-3T3 cells. In vitro, the CAAX peptidomimetic FTI-232 exhibits potent inhibition of FTase activity (IC50 = 150 nM) and its carboxyl-methylated counterpart, FTI-244, inhibits Ras processing in vivo. Treatment of NIH-3T3 cells with FTI-244 inhibits PDGF-induced DNA synthesis but not stimulation of mitogen-activated protein kinase (MAPK). However, FTI-244 significantly reduces PDGF-induced tyrosine phosphorylation levels of PDGF receptor (PDGFR) as well as its association with, and activation of, phosphatidylinositol-3-kinase (PI-3-K), a key enzyme in PDGF-induced mitogenesis.


Subject(s)
Alkyl and Aryl Transferases , Oligopeptides/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Protein Kinase Inhibitors , Receptors, Platelet-Derived Growth Factor/metabolism , Tyrosine/metabolism , 3T3 Cells , Animals , DNA Replication , Enzyme Activation , Farnesyltranstransferase , Mice , Molecular Mimicry , Phosphatidylinositol 3-Kinases , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Kinases/metabolism , Protein Processing, Post-Translational , Signal Transduction , Transferases/antagonists & inhibitors , Transferases/metabolism , ras Proteins/metabolism
17.
J Biol Chem ; 270(2): 660-4, 1995 Jan 13.
Article in English | MEDLINE | ID: mdl-7822292

ABSTRACT

Cysteine farnesylation of the carboxyl-terminal tetrapeptide CAAX (C = Cys, A = Leu, Ile, or Val, X = Met or Ser) of the oncogene product Ras is required for its malignant transformation activity. As a consequence farnesyltransferase (FTase), the enzyme responsible for this lipid modification, has become one of the most sought-after targets for anticancer drug development. We have recently designed peptide mimics of the COOH-terminal Cys-Val-Ile-Met of KB-Ras where the dipeptide Val-Ile was replaced by aminobenzoic acid derivatives. Although these peptidomimetics are potent inhibitors of FTase in vitro, they retain several undesirable peptide features that hamper their use in vivo. We report here the design, synthesis, and biological activity of the first non-peptide mimetics of CAAX where the tripeptide AAX was replaced by biphenyl derivatives. (R)-4-[N-(3-mercapto-2-aminopropyl)]amino-3'- carboxybiphenyl, where the cysteine is linked to the biphenyl derivative through a secondary amine, contains no amino acids, lacks peptidic features, and has no hydrolyzable bonds. This peptidomimetic is a potent inhibitor of FTase in vitro (IC50 = 50-150 nM) and disrupts Ras processing in whole cells. Furthermore, this non-peptide mimetic of CAAX is highly selective for FTase (666-fold) relative to the closely related geranylgeranyltransferase I. This selectivity is also respected in vivo since the processing of Ras but not the geranylgeranylated Rap1A was disrupted in whole cells. Structure activity relationship studies revealed that FTase recognition and inhibitory potency of CAAX peptidomimetics require free thiol and carboxylate groups separated by a hydrophobic moiety, and that precise positioning of these functional groups must correspond to that of the parent CAAX. The true CAAX peptidomimetic described in this manuscript has several desirable features for further development as a potential anticancer agent. It is not metabolically inactivated by FTase, does not require a pro-drug strategy for inhibition in vivo, and is selective for farnesylation relative to geranylgeranylation.


Subject(s)
Alkyl and Aryl Transferases , Molecular Mimicry , Oligopeptides/pharmacology , Protein Processing, Post-Translational , Transferases/antagonists & inhibitors , ras Proteins/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , GTP-Binding Proteins/metabolism , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Oligopeptides/chemistry , gamma-Glutamyltransferase/metabolism , rap GTP-Binding Proteins
18.
J Biol Chem ; 269(17): 12410-3, 1994 Apr 29.
Article in English | MEDLINE | ID: mdl-8175645

ABSTRACT

Cysteine farnesylation of the ras oncogene product, p21ras, on its carboxyl-terminal CA1A2X box (C = cysteine, A = aliphatic, and X = methionine or serine) is required for its transforming activity. p21ras farnesyltransferase (FTase), the enzyme responsible or this important posttranslational modification can be inhibited by simple CA1A2X peptides. We have synthesized a family of CA1A2X peptidomimetics where the central 2 aliphatic amino acids are replaced by a variety of spacer groups with different shapes and conformational characteristics to investigate the structural requirements of these inhibitors. The biological activities of CA1A2X peptidomimetics, where the dipeptide "A1A2" is replaced by 3- or 4-aminomethylbenzoic acid (AMBA) and 3- or 4-aminobenzoic acid (ABA), are evaluated in a p21ras FTase inhibitory assay. Peptidomimetics Cys-4-ABA-Met and Cys-3-AMBA-Met contain spacers that provide good distance correspondence of the carboxylate and ammonium separation with that of the parent KB p21ras tetrapeptide, Cys-Val-Ile-Met, and are as potent FTase inhibitors (IC50 values of 50 and 100 nM, respectively). In contrast, replacing the central dipeptide with 4-AMBA reduced FTase inhibitory activity by 17-fold whereas replacement by 3-ABA reduces inhibitory activity of the peptidomimetics by 43-fold. Cys-4-ABA-Met (IC50 = 50 nM) is 128 times more potent as a p21ras FTase inhibitor than Cys-3-ABA-Met (IC50 = 6400 nM), yet these two peptidomimetics differ only in the substitution pattern around the phenyl ring. These results coupled with computer modeling studies demonstrate that the interaction between FTase and the peptidomimetics requires precise structural and conformational characteristics; in particular, correct positioning of the Cys and Met must be respected. Furthermore, Cys-3-AMBA-Met and Cys-4-ABA-Met are true inhibitors of p21ras FTase since they are not farnesylated by this enzyme, in contrast to Cys-Val-Ile-Met, which inhibits the enzyme by acting as alternative substrate. Computer modeling studies of the potent FTase inhibitor Cys-4-ABA-Met show that a folded conformation, where the thiol and carboxylate groups are close, is not possible. Therefore a beta-turn conformation that would result in the simultaneous coordination of the Cys-thiol and Met-carboxylate to zinc ion is not important for inhibition of p21ras FTase, as previously suggested.


Subject(s)
Alkyl and Aryl Transferases , Oligopeptides/chemical synthesis , Transferases/antagonists & inhibitors , Amino Acid Sequence , Animals , Drug Design , Molecular Sequence Data , Oligopeptides/pharmacology , Protein Conformation , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...