Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 148
Filter
1.
Molecules ; 26(22)2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34833870

ABSTRACT

Peanut produces prenylated stilbenoids upon biotic stress. However, the role of these compounds against oxidative stress have not been thoroughly elucidated. To this end, the antioxidant capacity of extracts enriched in prenylated stilbenoids and derivatives was studied. To produce these extracts, hairy root cultures of peanut cultivars Hull, Tifrunner, and Georgia Green were co-treated with methyl jasmonate, cyclodextrin, hydrogen peroxide, and magnesium chloride and then the stilbenoids were extracted from the culture medium. Among the three cultivars, higher levels of the stilbenoid derivatives arachidin-1 and arachidin-6 were detected in cultivar Tifrunner. Upon reaction with 2,2-diphenyl-1picrylhydrazyl, extracts from cultivar Tifrunner showed the highest antioxidant capacity with an IC50 of 6.004 µg/mL. Furthermore, these extracts had significantly higher antioxidant capacity at 6.25 µg/mL and 3.125 µg/mL when compared to extracts from cultivars Hull and Georgia Green. The stilbenoid-rich extracts from peanut hairy roots show high antioxidant capacity and merit further study as potential nutraceuticals to promote human health.


Subject(s)
Arachis/metabolism , Oxidative Stress/physiology , Stilbenes/metabolism , Antioxidants/analysis , Antioxidants/pharmacology , Culture Media , Eicosanoic Acids , Fabaceae/metabolism , Humans , Plant Extracts/pharmacology , Plant Roots/metabolism , Protein Prenylation/physiology , Stilbenes/chemistry , Stilbenes/isolation & purification , Stress, Physiological/physiology
2.
Pharmacol Res ; 167: 105528, 2021 05.
Article in English | MEDLINE | ID: mdl-33667685

ABSTRACT

Geranylgeranyl diphosphate synthase (GGDPS), an enzyme in the isoprenoid biosynthetic pathway (IBP), produces the isoprenoid (geranylgeranyl pyrophosphate, GGPP) used in protein geranylgeranylation reactions. Our prior studies utilizing triazole bisphosphonate-based GGDPS inhibitors (GGSIs) have revealed that these agents represent a novel strategy by which to induce cancer cell death, including multiple myeloma and pancreatic cancer. Statins inhibit the rate-limiting enzyme in the IBP and potentiate the effects of GGSIs in vitro. The in vivo effects of combination therapy with statins and GGSIs have not been determined. Here we evaluated the effects of combining VSW1198, a novel GGSI, with a statin (lovastatin or pravastatin) in CD-1 mice. Twice-weekly dosing with VSW1198 at the previously established maximally tolerated dose in combination with a statin led to hepatotoxicity, while once-weekly VSW1198-based combinations were feasible. No abnormalities in kidney, spleen, brain or skeletal muscle were observed with combination therapy. Combination therapy disrupted protein geranylgeranylation in vivo. Evaluation of hepatic isoprenoid levels revealed decreased GGPP levels in the single drug groups and undetectable GGPP levels in the combination groups. Additional studies with combinations using 50% dose-reductions of either VSW1198 or lovastatin revealed minimal hepatotoxicity with expected on-target effects of diminished GGPP levels and disruption of protein geranylgeranylation. Combination statin/GGSI therapy significantly slowed tumor growth in a myeloma xenograft model. Collectively, these studies are the first to demonstrate that combination IBP inhibitor therapy alters isoprenoid levels and disrupts protein geranylgeranylation in vivo as well as slows tumor growth in a myeloma xenograft model, thus providing the framework for future clinical exploration.


Subject(s)
Biosynthetic Pathways/drug effects , Diterpenes/administration & dosage , Drug Delivery Systems/methods , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Protein Prenylation/drug effects , Terpenes/metabolism , Triazoles/administration & dosage , Animals , Biosynthetic Pathways/physiology , Cell Line, Tumor , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Diterpenes/toxicity , Drug Evaluation, Preclinical/methods , Drug Therapy, Combination , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/toxicity , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/metabolism , Female , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Lovastatin/administration & dosage , Lovastatin/toxicity , Mice , Mice, Inbred NOD , Mice, SCID , Pravastatin/administration & dosage , Pravastatin/toxicity , Protein Prenylation/physiology , Terpenes/antagonists & inhibitors , Triazoles/toxicity , Xenograft Model Antitumor Assays/methods
3.
Mol Neurobiol ; 57(5): 2220-2231, 2020 May.
Article in English | MEDLINE | ID: mdl-31989383

ABSTRACT

Mevalonate pathway inhibitors have been extensively studied for their roles in cholesterol depletion and for inhibiting the prenylation and activation of various proteins. Inhibition of protein prenylation has potential therapeutic uses against neurological disorders, like neural cancers, neurodegeneration, and neurotramatic lesions. Protection against neurodegeneration and promotion of neuronal regeneration is regulated in large part by Ras superfamily small guanosine triphosphatases (GTPases), particularly the Ras, Rho, and Rab subfamilies. These proteins are prenylated to target them to cellular membranes. Prenylation can be specifically inhibited through altering the function of enzymes of the mevalonate pathway necessary for isoprenoid production and attachment to target proteins to elicit a variety of effects on neural cells. However, this approach does not address how prenylation affects a specific protein. This review focuses on the regulation of small GTPase prenylation, the different techniques to inhibit prenylation, and how this inhibition has affected neural cell processes.


Subject(s)
GTP Phosphohydrolases/metabolism , Nerve Tissue Proteins/metabolism , Protein Prenylation/physiology , Acyl Coenzyme A/metabolism , Amino Acid Motifs/drug effects , Animals , Biosynthetic Pathways/drug effects , Cell Membrane/metabolism , Dimethylallyltranstransferase/metabolism , Enzyme Activation , Humans , Methylation , Mevalonic Acid/metabolism , Protein Binding , Terpenes/metabolism
4.
Biochemistry ; 58(30): 3232-3242, 2019 07 30.
Article in English | MEDLINE | ID: mdl-31251043

ABSTRACT

Indole prenyltransferases catalyze the prenylation of l-tryptophan (l-Trp) and other indoles to produce a diverse set of natural products in bacteria, fungi, and plants, many of which possess useful biological properties. Among this family of enzymes, CymD from Salinispora arenicola catalyzes the reverse N1 prenylation of l-Trp, an unusual reaction given the poor nucleophilicity of the indole nitrogen. CymD utilizes dimethylallyl diphosphate (DMAPP) as the prenyl donor, catalyzing the dissociation of the diphosphate leaving group followed by nucleophilic attack of the indole nitrogen at the tertiary carbon of the dimethylallyl cation. To better understand the structural basis of selective indole N-alkylation reactions in biology, we have determined the X-ray crystal structures of CymD, the CymD-l-Trp complex, and the CymD-l-Trp-DMSPP complex (DMSPP is dimethylallyl S-thiolodiphosphate, an unreactive analogue of DMAPP). The orientation of l-Trp with respect to DMSPP reveals how the active site contour of CymD serves as a template to direct the reverse prenylation of the indole nitrogen. Comparison to PriB, a C6 bacterial indole prenyltransferase, offers further insight regarding the structural basis of regioselective indole prenylation. Isothermal titration calorimetry measurements indicate a synergistic relationship between l-Trp and DMSPP binding. Finally, activity assays demonstrate the selectivity of CymD for l-Trp and indole as prenyl acceptors. Collectively, these data establish a foundation for understanding and engineering the regioselectivity of indole prenylation by members of the prenyltransferase protein family.


Subject(s)
Dimethylallyltranstransferase/chemistry , Protein Prenylation/physiology , Tryptophan/chemistry , Catalysis , Dimethylallyltranstransferase/metabolism , Micromonosporaceae/enzymology , Protein Structure, Secondary , Protein Structure, Tertiary , Tryptophan/metabolism , X-Ray Diffraction/methods
5.
Biochem Biophys Res Commun ; 495(1): 1249-1256, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29180010

ABSTRACT

Cardiolipin and phosphatidic acid-binding protein (CLPABP) controls the stability of the mRNA harboring an AU-rich element (ARE) in the 3' UTR with the help of the RNA stabilizer, human antigen R (HuR). Although CLPABP is localized on the mitochondrial surface as a large protein-RNA complex, its precise role is not yet known. Recently, CLPABP was identified as an N-myristoylated protein. Here, we demonstrate the effects of N-myristoylation on the functions of CLPABP. In the present study, compared to the wild-type protein that possessed the "MG" motif at the N-terminus for N-myristoylation, the mutant CLPABP protein that lacked N-myristoylation modification site was unstable. Furthermore, the expression of the G/A mutant of CLPABP, which lacked N-myristoylation site, induced morphological alterations in mitochondria. Because pleckstrin homology domain-deleted mutant, which was fused with the N-myristoylation site derived from intact CLPABP, could not colocalize with mitochondria, N-myristoylation of CLPABP was predicted to affect its stability onto the mitochondrial membrane rather than its subcellular localization.


Subject(s)
Lipid Metabolism/physiology , Lipid-Linked Proteins/metabolism , Myristic Acid/metabolism , Protein Prenylation/physiology , Subcellular Fractions/metabolism , Animals , COS Cells , Chlorocebus aethiops , HEK293 Cells , Humans
6.
Biomed Pharmacother ; 96: 489-496, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29032332

ABSTRACT

The high prevalence of end-stage renal disease emphasizes the failure to provide therapies to effectively prevent and/or reverse renal fibrosis. Therefore, the aim of this study was to evaluate the effect of long-term treatment with chaethomellic acid A (CAA), which selectively blocks Ha-Ras farnesylation, on renal mass reduction-induced renal fibrosis. Male Wistar rats were sham-operated (SO) or subjected to 5/6 renal mass reduction (RMR). One week after surgery, rats were placed in four experimental groups: SO:SO rats without treatment (n=13); SO+CAA: SO rats treated with CAA (n=13); RMR:RMR rats without treatment (n=14); and RMR+CAA:RMR rats treated with CAA (n=13). CAA was intraperitoneally administered in a dose of 0.23µg/kg three times a week for six months. Renal fibrosis was evaluated by two-dimensional ultrasonography and histopathological analysis. The kidneys of the RMR animals treated with CAA showed a significantly decrease in the medullary echogenicity (p<0.05) compared with the RMR rats that received no treatment. Glomerulosclerosis and arteriolosclerosis scores were significantly lower (p<0.001) in the RMR+CAA group when compared with the RMR group. There were no significant differences in interstitial fibrosis, interstitial inflammation and tubular dilatation scores between the RMR+CAA and RMR groups. These data suggest that CAA can be a potential future drug to attenuate the progression of chronic kidney disease.


Subject(s)
Arteriolosclerosis/diagnostic imaging , Disease Models, Animal , Glomerulosclerosis, Focal Segmental/diagnostic imaging , Renal Agents/therapeutic use , Renal Insufficiency, Chronic/diagnostic imaging , Animals , Arteriolosclerosis/drug therapy , Arteriolosclerosis/metabolism , Drug Administration Schedule , Genes, ras/drug effects , Genes, ras/physiology , Glomerulosclerosis, Focal Segmental/drug therapy , Glomerulosclerosis, Focal Segmental/metabolism , Male , Protein Prenylation/drug effects , Protein Prenylation/physiology , Rats , Rats, Wistar , Renal Agents/pharmacology , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/metabolism , Time Factors , Treatment Outcome
7.
Proc Natl Acad Sci U S A ; 113(44): E6766-E6775, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27791178

ABSTRACT

Farnesylation and carboxymethylation of KRAS4b (Kirsten rat sarcoma isoform 4b) are essential for its interaction with the plasma membrane where KRAS-mediated signaling events occur. Phosphodiesterase-δ (PDEδ) binds to KRAS4b and plays an important role in targeting it to cellular membranes. We solved structures of human farnesylated-methylated KRAS4b in complex with PDEδ in two different crystal forms. In these structures, the interaction is driven by the C-terminal amino acids together with the farnesylated and methylated C185 of KRAS4b that binds tightly in the central hydrophobic pocket present in PDEδ. In crystal form II, we see the full-length structure of farnesylated-methylated KRAS4b, including the hypervariable region. Crystal form I reveals structural details of farnesylated-methylated KRAS4b binding to PDEδ, and crystal form II suggests the potential binding mode of geranylgeranylated-methylated KRAS4b to PDEδ. We identified a 5-aa-long sequence motif (Lys-Ser-Lys-Thr-Lys) in KRAS4b that may enable PDEδ to bind both forms of prenylated KRAS4b. Structure and sequence analysis of various prenylated proteins that have been previously tested for binding to PDEδ provides a rationale for why some prenylated proteins, such as KRAS4a, RalA, RalB, and Rac1, do not bind to PDEδ. Comparison of all four available structures of PDEδ complexed with various prenylated proteins/peptides shows the presence of additional interactions due to a larger protein-protein interaction interface in KRAS4b-PDEδ complex. This interface might be exploited for designing an inhibitor with minimal off-target effects.


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases/chemistry , 3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Protein Interaction Domains and Motifs , Protein Prenylation/physiology , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/metabolism , 3',5'-Cyclic-GMP Phosphodiesterases/genetics , Amino Acid Sequence , Binding Sites , Cell Membrane/metabolism , Crystallography, X-Ray , Genes, ras , Humans , Methylation , Models, Molecular , Molecular Conformation , Mutation , Protein Binding/physiology , Proto-Oncogene Proteins p21(ras)/genetics , Sequence Analysis , rac1 GTP-Binding Protein/metabolism , ral GTP-Binding Proteins/metabolism
8.
Am J Pathol ; 186(6): 1454-65, 2016 06.
Article in English | MEDLINE | ID: mdl-27106761

ABSTRACT

G proteins play essential roles in regulating fetal lung development, and any defects in their expression or function (eg, activation or posttranslational modification) can lead to lung developmental malformation. Geranylgeranyl diphosphate synthase (GGPPS) can modulate protein prenylation that is required for protein membrane-anchoring and activation. Here, we report that GGPPS regulates fetal lung branching morphogenesis possibly through controlling K-Ras prenylation during fetal lung development. GGPPS was continuously expressed in lung epithelium throughout whole fetal lung development. Specific deletion of geranylgeranyl diphosphate synthase 1 (Ggps1) in lung epithelium during fetal lung development resulted in neonatal respiratory distress syndrome-like disease. The knockout mice died at postnatal day 1 of respiratory failure, and the lungs showed compensatory pneumonectasis, pulmonary atelectasis, and hyaline membranes. Subsequently, we proved that lung malformations in Ggps1-deficient mice resulted from the failure of fetal lung branching morphogenesis. Further investigation revealed Ggps1 deletion blocked K-Ras geranylgeranylation and extracellular signal-related kinase 1 or 2/mitogen-activated protein kinase signaling, which in turn disturbed fibroblast growth factor 10 regulation on fetal lung branching morphogenesis. Collectively, our data suggest that GGPPS is essential for maintaining fetal lung branching morphogenesis, which is possibly through regulating K-Ras prenylation.


Subject(s)
Farnesyltranstransferase/metabolism , Lung/embryology , Multienzyme Complexes/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Blotting, Western , Fetal Development , Fluorescent Antibody Technique , Immunohistochemistry , Immunoprecipitation , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Prenylation/physiology , Real-Time Polymerase Chain Reaction
9.
J Pathol ; 238(1): 109-19, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26434932

ABSTRACT

Loss of first-phase insulin secretion associated with ß cell dysfunction is an independent predictor of type 2 diabetes mellitus (T2DM) onset. Here we found that a critical enzyme involved in protein prenylation, geranylgeranyl pyrophosphate synthase (GGPPS), is required to maintain first-phase insulin secretion. GGPPS shows a biphasic expression pattern in islets of db/db mice during the progression of T2DM: GGPPS is increased during the insulin compensatory period, followed by a decrease during ß cell dysfunction. Ggpps deletion in ß cells results in typical T2DM ß cell dysfunction, with blunted glucose-stimulated insulin secretion and consequent insulin secretion insufficiency. However, the number and size of islets and insulin biosynthesis are unaltered. Transmission electron microscopy shows a reduced number of insulin granules adjacent to the cellular membrane, suggesting a defect in docked granule pool formation, while the reserve pool is unaffected. Ggpps ablation depletes GGPP and impairs Rab27A geranylgeranylation, which is responsible for the docked pool deficiency in Ggpps-null mice. Moreover, GGPPS re-expression or GGPP administration restore glucose-stimulated insulin secretion in Ggpps-null islets. These results suggest that GGPPS-controlled protein geranylgeranylation, which regulates formation of the insulin granule docked pool, is critical for ß cell function and insulin release during the development of T2DM.


Subject(s)
B-Lymphocytes/metabolism , Diabetes Mellitus, Type 2/metabolism , Farnesyltranstransferase/metabolism , Insulin/metabolism , Multienzyme Complexes/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Blotting, Western , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Protein Prenylation/physiology , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Secretory Vesicles/metabolism , Secretory Vesicles/ultrastructure , Transfection , rab27 GTP-Binding Proteins
10.
Proteomics ; 16(5): 759-82, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26683279

ABSTRACT

Covalent lipid modifications of proteins are crucial for regulation of cellular plasticity, since they affect the chemical and physical properties and therefore protein activity, localization, and stability. Most recently, lipid modifications on proteins are increasingly attracting important regulatory entities in diverse signaling events and diseases. In all cases, the lipid moiety of modified proteins is essential to allow water-soluble proteins to strongly interact with membranes or to induce structural changes in proteins that are critical for elemental processes such as respiration, transport, signal transduction, and motility. Until now, roughly about ten lipid modifications on different amino acid residues are described at the UniProtKB database and even well-known modifications are underrepresented. Thus, it is of fundamental importance to develop a better understanding of this emerging and so far under-investigated type of protein modification. Therefore, this review aims to give a comprehensive and detailed overview about enzymatic and nonenzymatic lipidation events, will report their role in cellular biology, discuss their relevancy for diseases, and describe so far available bioanalytical strategies to analyze this highly challenging type of modification.


Subject(s)
Lipid Metabolism/physiology , Lipoylation/physiology , Protein Prenylation/physiology , Proteins/metabolism , Humans , Lipids/chemistry
11.
Sci Rep ; 5: 15916, 2015 Nov 02.
Article in English | MEDLINE | ID: mdl-26522388

ABSTRACT

Prenylated proteins play key roles in several human diseases including cancer, atherosclerosis and Alzheimer's disease. KRAS4b, which is frequently mutated in pancreatic, colon and lung cancers, is processed by farnesylation, proteolytic cleavage and carboxymethylation at the C-terminus. Plasma membrane localization of KRAS4b requires this processing as does KRAS4b-dependent RAF kinase activation. Previous attempts to produce modified KRAS have relied on protein engineering approaches or in vitro farnesylation of bacterially expressed KRAS protein. The proteins produced by these methods do not accurately replicate the mature KRAS protein found in mammalian cells and the protein yield is typically low. We describe a protocol that yields 5-10 mg/L highly purified, farnesylated, and methylated KRAS4b from insect cells. Farnesylated and methylated KRAS4b is fully active in hydrolyzing GTP, binds RAF-RBD on lipid Nanodiscs and interacts with the known farnesyl-binding protein PDEδ.


Subject(s)
Lipids/physiology , Protein Prenylation/physiology , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Biophysics/methods , Cell Membrane/metabolism , Cells, Cultured , Guanosine Triphosphate/metabolism , Humans , Insecta/metabolism , Methylation , Protein Binding/physiology , raf Kinases/metabolism
12.
J Biol Chem ; 290(48): 28822-33, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26463208

ABSTRACT

Plasmalogen biosynthesis is regulated by modulating fatty acyl-CoA reductase 1 stability in a manner dependent on cellular plasmalogen level. However, physiological significance of the regulation of plasmalogen biosynthesis remains unknown. Here we show that elevation of the cellular plasmalogen level reduces cholesterol biosynthesis without affecting the isoprenylation of proteins such as Rab and Pex19p. Analysis of intermediate metabolites in cholesterol biosynthesis suggests that the first oxidative step in cholesterol biosynthesis catalyzed by squalene monooxygenase (SQLE), an important regulator downstream HMG-CoA reductase in cholesterol synthesis, is reduced by degradation of SQLE upon elevation of cellular plasmalogen level. By contrast, the defect of plasmalogen synthesis causes elevation of SQLE expression, resulting in the reduction of 2,3-epoxysqualene required for cholesterol synthesis, hence implying a novel physiological consequence of the regulation of plasmalogen biosynthesis.


Subject(s)
Cholesterol/biosynthesis , Homeostasis/physiology , Plasmalogens/biosynthesis , Animals , CHO Cells , Cholesterol/genetics , Cricetinae , Cricetulus , Gene Expression Regulation, Enzymologic/physiology , HEK293 Cells , HeLa Cells , Humans , Hydroxymethylglutaryl CoA Reductases/biosynthesis , Hydroxymethylglutaryl CoA Reductases/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Plasmalogens/genetics , Protein Prenylation/physiology , Squalene Monooxygenase/biosynthesis , Squalene Monooxygenase/genetics , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
13.
Vascul Pharmacol ; 67-69: 38-47, 2015.
Article in English | MEDLINE | ID: mdl-25849951

ABSTRACT

BACKGROUND: The long pentraxin PTX3 is an acute-phase multi-functional protein that might play both positive and detrimental effects under different pathophysiological conditions. We previously showed that statins down-regulate the release of PTX3 in human endothelial cells (ECs). The present study investigated the mechanism mediating this effect, its occurrence in other cells involved in atherogenesis, and whether it takes place in experimental atherosclerosis. METHODS AND RESULTS: We found that atorvastatin (1-5 µmol/L) decreased the production and release of PTX3 in human ECs through a post-transcriptional effect. Co-incubation with mevalonate or geranylgeranyl pyrophosphate prevented this effect. Direct blockade of geranylgeranyl transferase I by GGTI-286, treatment with the Rac inhibitor NSC23766 or silencing of the geranylgeranylated GTPase Rac2 by siRNA closely mimicked the action of atorvastatin. In contrast, inactivation of other geranylgeranylated proteins such as RhoA, RhoB, and RhoC or Rac1 did not affect PTX3 release. In addition, we found that atorvastatin also decreased PTX3 secretion in aortic SMCs through a mechanism likely dependent on protein geranylgeranylation, while no effect was observed in monocytes. Finally, we found that atherosclerotic lesions from cholesterol-fed rabbits treated with atorvastatin (2.5 mg/kg/day for 8 weeks) showed less immunoreactive PTX3 than lesions from control animals. CONCLUSIONS: Results suggest that statins may interfere with PTX3 expression in vascular cells via inhibition of protein geranylgeranylation. Since PTX3 is increasingly regarded as an important mediator of the inflammatory response underlying atherosclerosis and its complications, these results highlight the need for further studies of the role of PTX3 and its potential pharmacological modulation in cardiovascular disease.


Subject(s)
Atorvastatin/pharmacology , C-Reactive Protein/biosynthesis , Endothelial Cells/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Protein Prenylation/physiology , Serum Amyloid P-Component/biosynthesis , Animals , C-Reactive Protein/antagonists & inhibitors , Cells, Cultured , Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Protein Prenylation/drug effects , Rabbits , Serum Amyloid P-Component/antagonists & inhibitors
14.
PLoS One ; 10(3): e0120716, 2015.
Article in English | MEDLINE | ID: mdl-25768003

ABSTRACT

Protein prenylation is a widespread and highly conserved eukaryotic post-translational modification that endows proteins with the ability to reversibly attach to intracellular membranes. The dynamic interaction of prenylated proteins with intracellular membranes is essential for their signalling functions and is frequently deregulated in disease processes such as cancer. As a result, protein prenylation has been pharmacologically targeted by numerous drug discovery programs, albeit with limited success. To a large extent, this can be attributed to an insufficient understanding of the interplay of different protein prenyltransferases and the combinatorial diversity of the prenylatable sequence space. Here, we report a high-throughput, growth-based genetic selection assay in Saccharomyces cerevisiae based on the Ras Recruitment System which, for the first time, has allowed us to create a comprehensive map of prenylatable protein sequences in S. cerevisiae. We demonstrate that potential prenylatable space is sparsely (6.2%) occupied leaving room for creation of synthetic orthogonal prenylatable sequences. To experimentally demonstrate that, we used the developed platform to engineer mutant farnesyltransferases that efficiently prenylate substrate motives that are not recognised by endogenous protein prenyltransferases. These uncoupled mutants can now be used as starting points for the systematic engineering of the eukaryotic protein prenylation machinery.


Subject(s)
Farnesyltranstransferase/metabolism , Protein Engineering/methods , Protein Prenylation/genetics , Protein Prenylation/physiology , Saccharomyces cerevisiae/physiology , Cloning, Molecular/methods , Drug Discovery/methods , Farnesyltranstransferase/genetics , High-Throughput Nucleotide Sequencing/methods
15.
J Neurosci ; 34(50): 16726-38, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25505325

ABSTRACT

Aerobic animals constantly monitor and adapt to changes in O2 levels. The molecular mechanisms involved in sensing O2 are, however, incompletely understood. Previous studies showed that a hexacoordinated globin called GLB-5 tunes the dynamic range of O2-sensing neurons in natural C. elegans isolates, but is defective in the N2 lab reference strain (McGrath et al., 2009; Persson et al., 2009). GLB-5 enables a sharp behavioral switch when O2 changes between 21 and 17%. Here, we show that GLB-5 also confers rapid behavioral and cellular recovery from exposure to hypoxia. Hypoxia reconfigures O2-evoked Ca(2+) responses in the URX O2 sensors, and GLB-5 enables rapid recovery of these responses upon re-oxygenation. Forward genetic screens indicate that GLB-5's effects on O2 sensing require PDL-1, the C. elegans ortholog of mammalian PrBP/PDE6δ protein. In mammals, PDE6δ regulates the traffic and activity of prenylated proteins (Zhang et al., 2004; Norton et al., 2005). PDL-1 promotes localization of GCY-33 and GCY-35, atypical soluble guanylate cyclases that act as O2 sensors, to the dendritic endings of URX and BAG neurons, where they colocalize with GLB-5. Both GCY-33 and GCY-35 are predicted to be prenylated. Dendritic localization is not essential for GCY-35 to function as an O2 sensor, but disrupting pdl-1 alters the URX neuron's O2 response properties. Functional GLB-5 can restore dendritic localization of GCY-33 in pdl-1 mutants, suggesting GCY-33 and GLB-5 are in a complex. Our data suggest GLB-5 and the soluble guanylate cyclases operate in close proximity to sculpt O2 responses.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Dendrites/enzymology , Globins/physiology , Guanylate Cyclase/metabolism , Oxygen/metabolism , Programmed Cell Death 1 Receptor/physiology , Protein Prenylation/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Soluble Guanylyl Cyclase
16.
Biochem Biophys Res Commun ; 452(4): 967-73, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25223799

ABSTRACT

Protein prenylation is a post-translational modification where farnesyl or geranylgeranyl groups are enzymatically attached to a C-terminal cysteine residue. This modification is essential for the activity of small cellular GTPases, as it allows them to associate with intracellular membranes. Dissociated from membranes, prenylated proteins need to be transported through the aqueous cytoplasm by protein carriers that shield the hydrophobic anchor from the solvent. One such carrier is Rho GDP dissociation inhibitor (RhoGDI). Recently, it was shown that prenylated Rho proteins that are not associated with RhoGDI are subjected to proteolysis in the cell. We hypothesized that the role of RhoGDI might be not only to associate with prenylated proteins but also to regulate the prenylation process in the cell. This idea is supported by the fact that RhoGDI binds both unprenylated and prenylated Rho proteins with high affinity in vitro, and hence, these interactions may affect the kinetics of prenylation. We addressed this question experimentally and found that RhoGDI increased the catalytic efficiency of geranylgeranyl transferase-I in RhoA prenylation. Nevertheless, we did not observe formation of a ternary RhoGDI∗RhoA∗GGTase-I complex, indicating sequential operation of geranylgeranyltransferase-I and RhoGDI. Our results suggest that RhoGDI accelerates Rho prenylation by kinetically trapping the reaction product, thereby increasing the rate of product release.


Subject(s)
Alkyl and Aryl Transferases/metabolism , Fibroblasts/metabolism , Protein Prenylation/physiology , rho-Specific Guanine Nucleotide Dissociation Inhibitors/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Catalysis , Cell Line , Cricetinae
17.
Reprod Toxicol ; 46: 115-20, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24732207

ABSTRACT

The 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR) is the rate-limiting enzyme in the biosynthesis of cholesterol and isoprenoids, which are substrates required for post-translational modification of signalling proteins that can potentially regulate various aspects of embryonic development. The HMGCR transcripts are detectable during early embryogenesis in both invertebrates and vertebrates, which suggests a conserved developmental requirement for mevalonate derivatives. Consistently, recent animal and in vitro studies have yielded valuable insights into potential morphogenic parameters that are modulated by HMGCR activity. These developmental end-points include brain and craniofacial morphogenesis, PGC migration and survival, myocardial epithelial migration and fusion, EC migration and survival, and vascular stabilization. By providing a synthesis of these studies, we hope that this review will highlight the need to comprehensively examine the entire suite of developmental processes regulated by HMGCR.


Subject(s)
Growth/physiology , Hydroxymethylglutaryl CoA Reductases/physiology , Animals , Hydroxymethylglutaryl CoA Reductases/genetics , Morphogenesis/genetics , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , Protein Prenylation/genetics , Protein Prenylation/physiology , Signal Transduction/physiology
18.
Mol Neurobiol ; 50(1): 49-59, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24515839

ABSTRACT

Inhibitors of the mevalonate pathway, including the highly prescribed statins, reduce the production of cholesterol and isoprenoids such as geranylgeranyl pyrophosphates. The Rho family of small guanine triphosphatases (GTPases) requires isoprenylation, specifically geranylgeranylation, for activation. Because Rho GTPases are primary regulators of actin filament rearrangements required for process extension, neurite arborization, and synaptic plasticity, statins may affect cognition or recovery from nervous system injury. Here, we assessed how manipulating geranylgeranylation affects neurite initiation, elongation, and branching in neuroblastoma growth cones. Treatment with the statin, lovastatin (20 µM), decreased measures of neurite initiation by 17.0 to 19.0 % when a source of cholesterol was present and increased neurite branching by 4.03- to 9.54-fold (regardless of exogenous cholesterol). Neurite elongation was increased by treatment with lovastatin only in cholesterol-free culture conditions. Treatment with lovastatin decreased growth cone actin filament content by up to 24.3 %. In all cases, co-treatment with the prenylation precursor, geranylgeraniol (10 µM), reversed the effect of lovastatin. In a prior work, statin effects on outgrowth were linked to modulating the actin depolymerizing factor, cofilin. In our assays, treatment with lovastatin or geranylgeraniol decreased cofilin phosphorylation in whole cell lysates. However, lovastatin increased cofilin phosphorylation in cell bodies and decreased it in growth cones, indicating differential regulation in specific cell regions. Together, we interpret these data to suggest that protein geranylgeranylation likely regulates growth cone actin filament content and subsequent neurite outgrowth through mechanisms that also affect actin nucleation and polymerization.


Subject(s)
Cell Body/metabolism , Cofilin 1/metabolism , Growth Cones/metabolism , Neurites/metabolism , Protein Prenylation/physiology , Animals , Cell Body/drug effects , Cell Line, Tumor , Cell Shape/drug effects , Diterpenes/pharmacology , Growth Cones/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/pharmacology , Neurites/drug effects , Protein Prenylation/drug effects , Rats
19.
Neurosci Lett ; 564: 27-31, 2014 Apr 03.
Article in English | MEDLINE | ID: mdl-24486886

ABSTRACT

Isoprenylation is crucial step for activating many intracellular signaling. The present study examined whether inhibition of the protein isoprenylation could affect neuropathic pain in partial sciatic nerve-ligated mice. Intrathecal treatment with a geranylgeranyl transferase I inhibitor GGTI-2133, but not with a farnesyl transferase inhibitor FTI-277, dose-dependently blocked the thermal hyperalgesia in partial sciatic nerve-ligated mice. Intrathecal treatment with GGTI-2133 also attenuated the mechanical allodynia in partial sciatic nerve-ligated mice. Phosphorylated MARCKS expression was increased in the ipsilateral side of the spinal cord dorsal horn in partial sciatic nerve-ligated mice, and this increase was attenuated by GGTI-2133 but not by FTI-277. These results suggest that protein isoprenylation by geranylgeranyl transferase I is involved in the neuropathic pain.


Subject(s)
Neuralgia/metabolism , Protein Prenylation/physiology , Animals , Hyperalgesia/metabolism , Imidazoles/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Leucine/analogs & derivatives , Leucine/pharmacology , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred ICR , Myristoylated Alanine-Rich C Kinase Substrate , Naphthalenes/pharmacology , Neuralgia/etiology , Phosphorylation , Sciatic Nerve/injuries
20.
Mol Neurobiol ; 50(1): 177-85, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24390573

ABSTRACT

Protein prenylation is an important lipid posttranslational modification of proteins. It includes protein farnesylation and geranylgeranylation, in which the 15-carbon farnesyl pyrophosphate or 20-carbon geranylgeranyl pyrophosphate is attached to the C-terminus of target proteins, catalyzed by farnesyl transferase or geranylgeranyl transferases, respectively. Protein prenylation facilitates the anchoring of proteins into the cell membrane and mediates protein-protein interactions. Among numerous proteins that undergo prenylation, small GTPases represent the largest group of prenylated proteins. Small GTPases are involved in regulating a plethora of cellular functions including synaptic plasticity. The prenylation status of small GTPases determines the subcellular locations and functions of the proteins. Dysregulation or dysfunction of small GTPases leads to the development of different types of disorders. Emerging evidence indicates that prenylated proteins, in particular small GTPases, may play important roles in the pathogenesis of Alzheimer's disease. This review focuses on the prenylation of Ras and Rho subfamilies of small GTPases and its relation to synaptic plasticity and Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Neuronal Plasticity/physiology , Neurons/metabolism , Protein Prenylation/physiology , Animals , Humans , Protein Processing, Post-Translational , ras Proteins/metabolism , rho GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...