Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Front Immunol ; 12: 724991, 2021.
Article in English | MEDLINE | ID: mdl-34539662

ABSTRACT

Mevalonate kinase deficiency (MKD) is an autoinflammatory metabolic disorder characterized by life-long recurring episodes of fever and inflammation, often without clear cause. MKD is caused by bi-allelic pathogenic variants in the MVK gene, resulting in a decreased activity of the encoded enzyme mevalonate kinase (MK). MK is an essential enzyme in the isoprenoid biosynthesis pathway, which generates both non-sterol and sterol isoprenoids. The inflammatory symptoms of patients with MKD point to a major role for isoprenoids in the regulation of the innate immune system. In particular a temporary shortage of the non-sterol isoprenoid geranylgeranyl pyrophosphate (GGPP) is increasingly linked with inflammation in MKD. The shortage of GGPP compromises protein prenylation, which is thought to be one of the main causes leading to the inflammatory episodes in MKD. In this review, we discuss current views and the state of knowledge of the pathogenetic mechanisms in MKD, with particular focus on the role of compromised protein prenylation.


Subject(s)
Inflammation/immunology , Mevalonate Kinase Deficiency/genetics , Protein Prenylation/genetics , Terpenes/metabolism , Biosynthetic Pathways , Genetic Association Studies , Humans , Immunotherapy , Mevalonate Kinase Deficiency/diagnosis , Mevalonate Kinase Deficiency/drug therapy , Mevalonate Kinase Deficiency/metabolism
2.
Acta Neuropathol Commun ; 9(1): 129, 2021 07 27.
Article in English | MEDLINE | ID: mdl-34315531

ABSTRACT

The pathogenic mechanisms underlying the development of Alzheimer's disease (AD) remain elusive and to date there are no effective prevention or treatment for AD. Farnesyltransferase (FT) catalyzes a key posttranslational modification process called farnesylation, in which the isoprenoid farnesyl pyrophosphate is attached to target proteins, facilitating their membrane localization and their interactions with downstream effectors. Farnesylated proteins, including the Ras superfamily of small GTPases, are involved in regulating diverse physiological and pathological processes. Emerging evidence suggests that isoprenoids and farnesylated proteins may play an important role in the pathogenesis of AD. However, the dynamics of FT and protein farnesylation in human brains and the specific role of neuronal FT in the pathogenic progression of AD are not known. Here, using postmortem brain tissue from individuals with no cognitive impairment (NCI), mild cognitive impairment (MCI), or Alzheimer's dementia, we found that the levels of FT and membrane-associated H-Ras, an exclusively farnesylated protein, and its downstream effector ERK were markedly increased in AD and MCI compared with NCI. To elucidate the specific role of neuronal FT in AD pathogenesis, we generated the transgenic AD model APP/PS1 mice with forebrain neuron-specific FT knockout, followed by a battery of behavioral assessments, biochemical assays, and unbiased transcriptomic analysis. Our results showed that the neuronal FT deletion mitigates memory impairment and amyloid neuropathology in APP/PS1 mice through suppressing amyloid generation and reversing the pathogenic hyperactivation of mTORC1 signaling. These findings suggest that aberrant upregulation of protein farnesylation is an early driving force in the pathogenic cascade of AD and that targeting FT or its downstream signaling pathways presents a viable therapeutic strategy against AD.


Subject(s)
Alzheimer Disease/genetics , Brain/metabolism , Farnesyltranstransferase/genetics , Neurons/metabolism , Protein Prenylation/genetics , Aged, 80 and over , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Behavior, Animal , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases , Farnesyltranstransferase/metabolism , Female , Humans , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Plaque, Amyloid/genetics , Plaque, Amyloid/metabolism , Presenilin-1/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction
3.
J Genet Genomics ; 48(4): 300-311, 2021 04 20.
Article in English | MEDLINE | ID: mdl-34049800

ABSTRACT

Vascular development is essential for the establishment of the circulatory system during embryonic development and requires the proliferation of endothelial cells. However, the underpinning regulatory mechanisms are not well understood. Here, we report that geranylgeranyl pyrophosphate (GGPP), a metabolite involved in protein geranylgeranylation, plays an indispensable role in embryonic vascular development. GGPP is synthesized by geranylgeranyl pyrophosphate synthase (GGPPS) in the mevalonate pathway. The selective knockout of Ggpps in endothelial cells led to aberrant vascular development and embryonic lethality, resulting from the decreased proliferation and enhanced apoptosis of endothelial cells during vasculogenesis. The defect in protein geranylgeranylation induced by GGPP depletion inhibited the membrane localization of RhoA and enhanced yes-associated protein (YAP) phosphorylation, thereby prohibiting the entry of YAP into the nucleus and the expression of YAP target genes related to cell proliferation and the antiapoptosis process. Moreover, inhibition of the mevalonate pathway by simvastatin induced endothelial cell proliferation defects and apoptosis, which were ameliorated by GGPP. Geranylgeraniol (GGOH), a precursor of GGPP, ameliorated the harmful effects of simvastatin on vascular development of developing fetuses in pregnant mice. These results indicate that GGPP-mediated protein geranylgeranylation is essential for endothelial cell proliferation and the antiapoptosis process during embryonic vascular development.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Embryonic Development/genetics , Farnesyltranstransferase/genetics , Multienzyme Complexes/genetics , Animals , Cell Differentiation/drug effects , Embryo, Mammalian , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Mice , Morphogenesis/genetics , Polyisoprenyl Phosphates/metabolism , Polyisoprenyl Phosphates/pharmacology , Pregnancy , Protein Prenylation/drug effects , Protein Prenylation/genetics , YAP-Signaling Proteins/genetics , rhoA GTP-Binding Protein/genetics
4.
Horm Metab Res ; 53(2): 124-131, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33307558

ABSTRACT

3-Hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR) is the rate-limiting enzyme of the mevalonate pathway, which generates cholesterol and non-sterol compounds such as isoprenoid, which are involved in key steps of tumorigenesis such as cell growth and proliferation. Our aim was to evaluate the role of the mevalonate pathway in adrenocortical tumors (ACTs). Expression pattern of HMGCR, FDFT1, LDLR, SCARB1, StAR, TSPO, CYP11A1, CYP11B1, CYP17A1, CYP21A1, and HSD3B1 genes, involved in the mevalonate pathway and steroidogenesis, was quantified by real-time RT-PCR in 46 ACT [14 adenomas (ACA) and 11 carcinomas (ACC) from adults and 13 ACA and 8 ACC from pediatric patients]. Effects of the mevalonate pathway inhibition on NCI-H295A cell viability was assessed by colorimetric assay. HMGCR was overexpressed in most adult ACT. The expression of TSPO, STAR, CYP11B1, CYP21A1, and HSD3B1 in adult ACC was significantly lower than in ACA (p<0.05). Regarding pediatric ACT, the expression of genes involved in steroidogenesis was not different between ACA and ACC. Inhibition of isoprenoid production significantly decreased the viability of NCI-H295A cells (p<0.05). However, cholesterol synthesis blockage did not show the same effect on cell viability. Low expression of TSPO ,: StAR, CYP11B1, CYP21A1, and HSD3B1 characterized a signature of adult ACCs. Our data suggest that HMGCR overexpression in adult ACC might lead to intracellular isoprenoid accumulation and cell proliferation. Therefore, the mevalonate pathway is a potential target for ACC treatment.


Subject(s)
Adrenal Cortex Neoplasms/metabolism , Adrenal Cortex Neoplasms/pathology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Metabolic Networks and Pathways , Mevalonic Acid/metabolism , Adolescent , Adrenal Cortex Neoplasms/genetics , Adult , Aged , Cell Line, Tumor , Cell Survival/genetics , Child, Preschool , Cholesterol/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Infant , Male , Metabolic Networks and Pathways/genetics , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Protein Prenylation/genetics , Steroids/biosynthesis , Young Adult
5.
Cells ; 9(11)2020 11 20.
Article in English | MEDLINE | ID: mdl-33233551

ABSTRACT

Nuclear shape modulates cell behavior and function, while aberrant nuclear morphologies correlate with pathological phenotype severity. Nevertheless, functions of specific nuclear morphological features and underlying molecular mechanisms remain poorly understood. Here, we investigate a nucleus-intrinsic mechanism driving nuclear lobulation and segmentation concurrent with granulocyte specification, independently from extracellular forces and cytosolic cytoskeleton contributions. Transcriptomic regulation of cholesterol biosynthesis is equally concurrent with nuclear remodeling. Its putative role as a regulatory element is supported by morphological aberrations observed upon pharmacological impairment of several enzymatic steps of the pathway, most prominently the sterol ∆14-reductase activity of laminB-receptor and protein prenylation. Thus, we support the hypothesis of a nuclear-intrinsic mechanism for nuclear shape control with the putative involvement of the recently discovered GGTase III complex. Such process could be independent from or complementary to the better studied cytoskeleton-based nuclear remodeling essential for cell migration in both physiological and pathological contexts such as immune system function and cancer metastasis.


Subject(s)
Cytoskeleton/metabolism , Granulocytes/metabolism , Protein Prenylation/genetics , HL-60 Cells , Humans , Models, Molecular
6.
J Exp Med ; 217(2)2020 02 03.
Article in English | MEDLINE | ID: mdl-31722972

ABSTRACT

Thymocyte egress is a critical determinant of T cell homeostasis and adaptive immunity. Despite the roles of G protein-coupled receptors in thymocyte emigration, the downstream signaling mechanism remains poorly defined. Here, we report the discrete roles for the two branches of mevalonate metabolism-fueled protein prenylation pathway in thymocyte egress and immune homeostasis. The protein geranylgeranyltransferase Pggt1b is up-regulated in single-positive thymocytes, and loss of Pggt1b leads to marked defects in thymocyte egress and T cell lymphopenia in peripheral lymphoid organs in vivo. Mechanistically, Pggt1b bridges sphingosine-1-phosphate and chemokine-induced migratory signals with the activation of Cdc42 and Pak signaling and mevalonate-dependent thymocyte trafficking. In contrast, the farnesyltransferase Fntb, which mediates a biochemically similar process of protein farnesylation, is dispensable for thymocyte egress but contributes to peripheral T cell homeostasis. Collectively, our studies establish context-dependent effects of protein prenylation and unique roles of geranylgeranylation in thymic egress and highlight that the interplay between cellular metabolism and posttranslational modification underlies immune homeostasis.


Subject(s)
Alkyl and Aryl Transferases/metabolism , Mevalonic Acid/metabolism , Protein Prenylation/genetics , Thymocytes/metabolism , Alkyl and Aryl Transferases/genetics , Animals , Cell Movement/genetics , Cells, Cultured , Farnesyltranstransferase/genetics , Farnesyltranstransferase/metabolism , Homeostasis/genetics , Homeostasis/immunology , Lymphopenia/genetics , Lysophospholipids/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics , Sphingosine/analogs & derivatives , Sphingosine/metabolism , T-Lymphocytes/metabolism , Thymus Gland/immunology , cdc42 GTP-Binding Protein/metabolism , p21-Activated Kinases/metabolism
7.
Front Immunol ; 10: 1900, 2019.
Article in English | MEDLINE | ID: mdl-31474985

ABSTRACT

The rare autoinflammatory disease mevalonate kinase deficiency (MKD, which includes HIDS and mevalonic aciduria) is caused by recessive, pathogenic variants in the MVK gene encoding mevalonate kinase. Deficiency of this enzyme decreases the synthesis of isoprenoid lipids and thus prevents the normal post-translational prenylation of small GTPase proteins, which then accumulate in their unprenylated form. We recently optimized a sensitive assay capable of detecting unprenylated Rab GTPase proteins in peripheral blood mononuclear cells (PBMCs) and showed that this assay distinguished MKD from other autoinflammatory diseases. We have now analyzed PBMCs from an additional six patients with genetically-confirmed MKD (with different compound heterozygous MVK genotypes), and compared these with PBMCs from three healthy volunteers and four unaffected control individuals heterozygous for the commonest pathogenic variant, MVKV377I . We detected a clear accumulation of unprenylated Rab proteins, as well as unprenylated Rap1A by western blotting, in all six genetically-confirmed MKD patients compared to heterozygous controls and healthy volunteers. Furthermore, in the three subjects for whom measurements of residual mevalonate kinase activity was available, enzymatic activity inversely correlated with the extent of the defect in protein prenylation. Finally, a heterozygous MVKV377I patient presenting with autoinflammatory symptoms did not have defective prenylation, indicating a different cause of disease. These findings support the notion that the extent of loss of enzyme function caused by biallelic MVK variants determines the severity of defective protein prenylation, and the accumulation of unprenylated proteins in PBMCs may be a sensitive and consistent biomarker that could be used to aid, or help rule out, diagnosis of MKD.


Subject(s)
Leukocytes, Mononuclear/metabolism , Mevalonate Kinase Deficiency/genetics , Mutation , Phosphotransferases (Alcohol Group Acceptor)/genetics , Adult , Biomarkers/metabolism , Cells, Cultured , Child , Female , Genotype , Hereditary Autoinflammatory Diseases/diagnosis , Hereditary Autoinflammatory Diseases/genetics , Hereditary Autoinflammatory Diseases/metabolism , Humans , Male , Mevalonate Kinase Deficiency/diagnosis , Mevalonate Kinase Deficiency/metabolism , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Prenylation/genetics , rap1 GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/metabolism
8.
Sci Rep ; 9(1): 7615, 2019 05 20.
Article in English | MEDLINE | ID: mdl-31110208

ABSTRACT

There is still no successful strategy to treat Huntington's disease, an inherited autosomal disorder associated with the aggregation of mutated forms of the huntingtin protein containing polyglutamine tracts with more than 36 repeats. Recent experimental evidence is challenging the conventional view of the disease by revealing transcellular transfer of mutated huntingtin proteins which are able to seed oligomers involving wild type forms of the protein. Here we decipher the molecular mechanism of this unconventional heterogeneous oligomerization by performing discrete molecular dynamics simulations. We identify the most probable oligomer conformations and the molecular regions that can be targeted to destabilize them. Our computational findings are complemented experimentally by fluorescence-lifetime imaging microscopy/fluorescence resonance energy transfer (FLIM-FRET) of cells co-transfected with huntingtin proteins containing short and large polyglutamine tracts. Our work clarifies the structural features responsible for heterogeneous huntingtin aggregation with possible implications to contrast the prion-like spreading of Huntington's disease.


Subject(s)
Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Humans , Huntington Disease/genetics , Huntington Disease/metabolism , Mutation/genetics , Peptides/metabolism , Protein Prenylation/genetics , Transfection/methods
9.
Genetics ; 210(4): 1301-1316, 2018 12.
Article in English | MEDLINE | ID: mdl-30257935

ABSTRACT

Protein isoprenylation targets a subset of COOH-terminal Cxxx tetrapeptide sequences that has been operationally defined as a CaaX motif. The specificity of the farnesyl transferase toward each of the possible 8000 combinations of Cxxx sequences, however, remains largely unresolved. In part, it has been difficult to consolidate results stemming from in vitro and in silico approaches that yield a wider array of prenylatable sequences relative to those known in vivo We have investigated whether this disconnect results from the multistep complexity of post-translational modification that occurs in vivo to CaaX proteins. For example, the Ras GTPases undergo isoprenylation followed by additional proteolysis and carboxymethylation events at the COOH-terminus. By contrast, Saccharomyces cerevisiae Hsp40 Ydj1p is isoprenylated but not subject to additional modification. In fact, additional modifications are detrimental to Ydj1p activity in vivo We have taken advantage of the properties of Ydj1p and a Ydj1p-dependent growth assay to identify sequences that permit Ydj1p isoprenylation in vivo while simultaneously selecting against nonprenylatable and more extensively modified sequences. The recovered sequences are largely nonoverlapping with those previously identified using an in vivo Ras-based yeast reporter. Moreover, most of the sequences are not readily predicted as isoprenylation targets by existing prediction algorithms. Our results reveal that the yeast CaaX-type prenyltransferases can utilize a range of sequence combinations that extend beyond the traditional constraints for CaaX proteins, which implies that more proteins may be isoprenylated than previously considered.


Subject(s)
Alkyl and Aryl Transferases/genetics , HSP40 Heat-Shock Proteins/genetics , Protein Prenylation/genetics , Saccharomyces cerevisiae Proteins/genetics , Amino Acid Motifs/genetics , Amino Acid Sequence/genetics , Protein Processing, Post-Translational/genetics , Saccharomyces cerevisiae/genetics , ras Proteins/genetics
10.
Curr Genet ; 64(2): 341-344, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28929213

ABSTRACT

Isoprenylcysteine-O-Carboxyl Methyltransferase (ICMT) catalyzes the final step in the prenylation process of different proteins including members of the Ras superfamily of GTPases. While cysteine methylation is essential in mammalian cells for growth, membrane association, and signalling by Ras and Rho GTPases, its role during signal transduction events in simple eukaryotes like yeasts appears irrelevant. By using a multidisciplinary approach our group has recently shown that, contrary to this initial assumption, in the fission yeast Schizosaccharomyces pombe ICMT activity encoded by the Mam4 gene is not only important to promote selective plasma membrane targeting of Ras and specific Rho GTPases, but also to allow precise downstream signalling to the mitogen-activated protein kinase and target of rapamycin pathways in response to diverse environmental cues. Thus, the dynamic regulation of in vivo methylation as a modulator of GTPase localization and function is an evolutionary conserved mechanism, making fission yeast an appealing model organism to study the regulation of this process.


Subject(s)
Cysteine/metabolism , Protein Methyltransferases/genetics , Protein Processing, Post-Translational/genetics , rho GTP-Binding Proteins/genetics , Cell Membrane/genetics , Cell Membrane/metabolism , Cysteine/genetics , Methylation , Protein Prenylation/genetics , Schizosaccharomyces/genetics , Signal Transduction/genetics
11.
Expert Rev Proteomics ; 14(6): 515-528, 2017 06.
Article in English | MEDLINE | ID: mdl-28521569

ABSTRACT

INTRODUCTION: Protein prenylation is a ubiquitous covalent post-translational modification characterized by the addition of farnesyl or geranylgeranyl isoprenoid groups to a cysteine residue located near the carboxyl terminal of a protein. It is essential for the proper localization and cellular activity of numerous proteins, including Ras family GTPases and G-proteins. In addition to its roles in cellular physiology, the prenylation process has important implications in human diseases and in the recent years, it has become attractive target of inhibitors with therapeutic potential. Areas covered: This review attempts to summarize the basic aspects of prenylation integrating them with biological functions in diseases and giving an account of the current status of prenylation inhibitors as potential therapeutics. We also summarize the methodologies for the characterization of this modification. Expert commentary: The growing body of evidence suggesting an important role of prenylation in diseases and the subsequent development of inhibitors of the enzymes responsible for this modification lead to the urgent need to identify the full spectrum of prenylated proteins that are altered in the disease or affected by drugs. Proteomic tools to analyze prenylated proteins are recently emerging, thanks to the advancement in the field of mass spectrometry coupled to enrichment strategies.


Subject(s)
Protein Prenylation/genetics , Protein Processing, Post-Translational/genetics , Proteins/genetics , Proteomics , Cysteine/genetics , Humans
12.
J Cell Sci ; 129(9): 1781-91, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27034135

ABSTRACT

There are roughly 14 distinct heritable autosomal dominant diseases associated with mutations in lamins A/C, including Emery-Dreifuss muscular dystrophy (EDMD). The mechanical model proposes that the lamin mutations change the mechanical properties of muscle nuclei, leading to cell death and tissue deterioration. Here, we developed an experimental protocol that analyzes the effect of disease-linked lamin mutations on the response of nuclei to mechanical strain in living Caenorhabditis elegans We found that the EDMD mutation L535P disrupts the nuclear mechanical response specifically in muscle nuclei. Inhibiting lamin prenylation rescued the mechanical response of the EDMD nuclei, reversed the muscle phenotypes and led to normal motility. The LINC complex and emerin were also required to regulate the mechanical response of C. elegans nuclei. This study provides evidence to support the mechanical model and offers a potential future therapeutic approach towards curing EDMD.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Lamins , Models, Biological , Movement , Muscular Dystrophy, Emery-Dreifuss , Mutation, Missense , Nuclear Proteins , Phenotype , Amino Acid Substitution , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Cycle Proteins , Cell Nucleus/genetics , Cell Nucleus/metabolism , Disease Models, Animal , Lamins/genetics , Lamins/metabolism , Muscular Dystrophy, Emery-Dreifuss/genetics , Muscular Dystrophy, Emery-Dreifuss/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Prenylation/genetics
13.
Hum Mol Genet ; 25(10): 2031-2044, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26936825

ABSTRACT

The small GTPase, ADP-ribosylation factor-like 3 (ARL3), has been proposed to participate in the transport of proteins in photoreceptor cells. Moreover, it has been implicated in the pathogenesis associated with X-linked retinitis pigmentosa (XLRP) resulting from mutations in the ARL3 GTPase activating protein, retinitis pigmentosa 2 (RP2). To determine the importance of ARL3 in rod photoreceptor cells, we generated transgenic mice expressing a dominant active form of ARL3 (ARL3-Q71L) under a rod-specific promoter. ARL3-Q71L animals exhibited extensive rod cell death after post-natal day 30 (PN30) and degeneration was complete by PN70. Prior to the onset of cell death, rod photoresponse was significantly reduced along with a robust decrease in rod phosphodiesterase 6 (PDE6) and G-protein receptor kinase-1 (GRK1) levels. Furthermore, assembled phosphodiesterase-6 (PDE6) subunits, rod transducin and G-protein receptor kinase-1 (GRK1) accumulated on large punctate structures within the inner segment in ARL3-Q71L retina. Defective trafficking of prenylated proteins is likely due to sequestration of prenyl binding protein δ (PrBPδ) by ARL3-Q71L as we demonstrate a specific interaction between these proteins in the retina. Unexpectedly, our studies also revealed a novel role for ARL3 in the migration of photoreceptor nuclei. In conclusion, this study identifies ARL3 as a key player in prenylated protein trafficking in rod photoreceptor cells and establishes the potential role for ARL3 dysregulation in the pathogenesis of RP2-related forms of XLRP.


Subject(s)
ADP-Ribosylation Factors/genetics , Genetic Diseases, X-Linked/genetics , Pyrophosphatases/genetics , Retinitis Pigmentosa/genetics , ADP-Ribosylation Factors/biosynthesis , Animals , Cyclic Nucleotide Phosphodiesterases, Type 6/biosynthesis , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Eye Proteins/biosynthesis , Eye Proteins/genetics , G-Protein-Coupled Receptor Kinase 1/biosynthesis , G-Protein-Coupled Receptor Kinase 1/genetics , GTP-Binding Proteins , Gene Expression Regulation , Genetic Diseases, X-Linked/pathology , Humans , Membrane Proteins , Mice , Mice, Transgenic , Protein Prenylation/genetics , Retina/metabolism , Retina/pathology , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Retinitis Pigmentosa/pathology , Rod Cell Outer Segment/metabolism , Rod Cell Outer Segment/pathology
14.
Cell Death Dis ; 6: e1825, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26181205

ABSTRACT

Reduction of mitochondrial DNA (mtDNA) content induces the reduction of oxidative phosphorylation and dependence on fermentative glycolysis, that is, the Warburg effect. In aggressive prostate cancer (PCa), the reduction of mtDNA reduces oxygen consumption, increases intracellular oxygen concentration, and induces constitutive activation of Ras. Many essential proteins for cell death, growth, differentiation, and development, such as Ras, require prenylation for subcellular localization and activation. Prenylation of a protein is defined as the attachment of isoprenoids to a cysteine residue at or near the C-terminus. 3-Hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGR) produces isoprenoids, and is posttranslationally regulated by oxygen. We investigated a critical role of intracellular oxygen in membrane localization of prenylated proteins. Localization of prenylated proteins (H-Ras, prelamin A/C, and Rab5a) was observed in poorly differentiated PCa (PC-3) and well-differentiated PCa (LNCaP) cells. PC-3 cells exhibited high intracellular oxygen concentration, and H-Ras, prelamin A/C, and Rab5a were localized to various membranes (Golgi and plasma membrane, nuclear membrane, and early endosomes, respectively). Remarkably, exogenous hypoxia (0.2% O2) in PC-3 cells induced intracellular hypoxia and changed the localization of the prenylated proteins. H-Ras and Rab5a were translocated to cytosol, and prelamin A/C was in the nucleus forming an abnormal nuclear envelope. The localization was reversed by mevalonate indicating the involvement of mevalonate pathway. In contrast, in LNCaP cells, exhibiting low intracellular oxygen concentration, H-Ras and Rab5a were localized in the cytosol, and prelamin A/C was inside the nucleus forming an inadequate nuclear envelope. Exogenous hyperoxia (40% O2) increased the intracellular oxygen concentration and induced Ras translocation from cytosol to the membrane. Prelamin A/C was translocated to the nuclear membrane and formed a proper nuclear envelope. Rab5a was translocated to the early endosomes. The specific localizations of the prenylated proteins were dependent on intracellular oxygen concentration. These results demonstrate that intracellular oxygen concentration regulates the localization and activation of prenylated proteins.


Subject(s)
Oxygen/metabolism , Prostatic Neoplasms/metabolism , Protein Prenylation/genetics , rab5 GTP-Binding Proteins/genetics , Cell Line, Tumor , DNA, Mitochondrial/metabolism , Endosomes/genetics , Endosomes/metabolism , Humans , Hydroxymethylglutaryl CoA Reductases , Male , Oxidative Phosphorylation , Oxygen Consumption/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology
15.
J Cell Biol ; 208(7): 881-96, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25825516

ABSTRACT

Kinetochore (KT) localization of mitotic checkpoint proteins is essential for their function during mitosis. hSpindly KT localization is dependent on the RZZ complex and hSpindly recruits the dynein-dynactin complex to KTs during mitosis, but the mechanism of hSpindly KT recruitment is unknown. Through domain-mapping studies we characterized the KT localization domain of hSpindly and discovered it undergoes farnesylation at the C-terminal cysteine residue. The N-terminal 293 residues of hSpindly are dispensable for its KT localization. Inhibition of farnesylation using a farnesyl transferase inhibitor (FTI) abrogated hSpindly KT localization without affecting RZZ complex, CENP-E, and CENP-F KT localization. We showed that hSpindly is farnesylated in vivo and farnesylation is essential for its interaction with the RZZ complex and hence KT localization. FTI treatment and hSpindly knockdown displayed the same mitotic phenotypes, indicating that hSpindly is a key FTI target in mitosis. Our data show a novel role of lipidation in targeting a checkpoint protein to KTs through protein-protein interaction.


Subject(s)
Carrier Proteins/genetics , Chromosome Segregation/genetics , Kinetochores/metabolism , M Phase Cell Cycle Checkpoints/genetics , Protein Prenylation/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Farnesyltranstransferase/antagonists & inhibitors , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/genetics , Mutation , Protein Structure, Tertiary , RNA Interference , RNA, Small Interfering , Sequence Deletion , Spindle Apparatus/genetics
16.
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
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.
Biomed Res Int ; 2013: 416534, 2013.
Article in English | MEDLINE | ID: mdl-23936796

ABSTRACT

Inhibition of PMPMEase, a key enzyme in the polyisoprenylation pathway, induces cancer cell death. In this study, purified PMPMEase was inhibited by the chemopreventive agent, curcumin, with a K(i) of 0.3 µM (IC50 = 12.4 µM). Preincubation of PMPMEase with 1 mM curcumin followed by gel-filtration chromatography resulted in recovery of the enzyme activity, indicative of reversible inhibition. Kinetics analysis with N-para-nitrobenzoyl-S-trans,trans-farnesylcysteine methyl ester substrate yielded K M values of 23.6 ± 2.7 and 85.3 ± 15.3 µM in the absence or presence of 20 µM curcumin, respectively. Treatment of colorectal cancer (Caco2) cells with curcumin resulted in concentration-dependent cell death with an EC50 of 22.0 µg/mL. PMPMEase activity in the curcumin-treated cell lysate followed a similar concentration-dependent profile with IC50 of 22.6 µg/mL. In colorectal cancer tissue microarray studies, PMPMEase immunoreactivity was significantly higher in 88.6% of cases compared to normal colon tissues (P < 0.0001). The mean scores ± SEM were 91.7 ± 11.4 (normal), 75.0 ± 14.4 (normal adjacent), 294.8 ± 7.8 (adenocarcinoma), and 310.0 ± 22.6 (mucinous adenocarcinoma), respectively. PMPMEase overexpression in colorectal cancer and cancer cell death stemming from its inhibition is an indication of its possible role in cancer progression and a target for chemopreventive agents.


Subject(s)
Carboxylic Ester Hydrolases/metabolism , Colorectal Neoplasms/enzymology , Curcumin/administration & dosage , Protein Prenylation/genetics , Caco-2 Cells , Carboxylic Ester Hydrolases/chemistry , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Kinetics , Molecular Targeted Therapy
19.
J Biol Chem ; 288(38): 27444-27455, 2013 Sep 20.
Article in English | MEDLINE | ID: mdl-23908355

ABSTRACT

Mammalian cells can use exogenous isoprenols to generate isoprenoid diphosphate substrates for protein isoprenylation, but the mechanism, efficiency, and biological importance of this process are not known. We developed mass spectrometry-based methods using chemical probes and newly synthesized stable isotope-labeled tracers to quantitate incorporation of exogenously provided farnesol, geranylgeraniol, and unnatural analogs of these isoprenols containing an aniline group into isoprenoid diphosphates and protein isoprenylcysteines by cultured human cancer cell lines. We found that at exogenous isoprenol concentrations >10 µM, this process can generate as much as 50% of the cellular isoprenoid diphosphate pool used for protein isoprenylation. Mutational activation of p53 in MDA-MB-231 breast cancer cells up-regulates the mevalonate pathway to promote tumor invasiveness. p53 silencing or pharmacological inhibition of HMG-CoA reductase in these cells decreases protein isoprenylation from endogenously synthesized isoprenoids but enhances the use of exogenous isoprenols for this purpose, indicating that this latter process is regulated independently of the mevalonate pathway. Our observations suggest unique opportunities for design of cancer cell-directed therapies and may provide insights into mechanisms underlying pleiotropic therapeutic benefits and unwanted side effects of mevalonate pathway inhibition.


Subject(s)
Antineoplastic Agents/pharmacology , Diterpenes/pharmacology , Farnesol/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mevalonic Acid/metabolism , Neoplasms/drug therapy , Polyisoprenyl Phosphates/metabolism , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Diterpenes/pharmacokinetics , Farnesol/analogs & derivatives , Farnesol/pharmacokinetics , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl CoA Reductases/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Protein Prenylation/drug effects , Protein Prenylation/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
20.
Insect Biochem Mol Biol ; 43(10): 947-58, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23907071

ABSTRACT

Geranylgeranyl diphosphate synthase (GGPPS) catalyzes the condensation of the non-allylic diphosphate, isopentenyl diphosphate (IPP; C5), with allylic diphosphates to generate the C20 prenyl chain (GGPP) used for protein prenylation and diterpenoid biosynthesis. Here, we cloned the cDNA of a GGPPS from the spruce budworm, Choristoneura fumiferana, and characterized the corresponding recombinant protein (rCfGGPPS). As shown for other type-III GGPPSs, rCfGGPPS preferred farnesyl diphosphate (FPP; C15) over other allylic substrates for coupling with IPP. Unexpectedly, rCfGGPPS displayed inhibition by its FPP substrate at low IPP concentration, suggesting the existence of a mechanism that may regulate intracellular FPP pools. rCfGGPPS was also inhibited by its product, GGPP, in a competitive manner with respect to FPP, as reported for human and bovine brain GGPPSs. A homology model of CfGGPPS was prepared and compared to human and yeast GGPPSs. Consistent with its enzymological properties, CfGGPPS displayed a larger active site cavity that can accommodate the binding of FPP and GGPP in the region normally occupied by IPP and the allylic isoprenoid tail, and the binding of GGPP in an alternate orientation seen for GGPP binding to the human protein. To begin exploring the role of CfGGPPS in protein prenylation, its transcripts were quantified by qPCR in whole insects, along with those of other genes involved in this pathway. CfGGPPS was expressed throughout insect development and the abundance of its transcripts covaried with that of other prenylation-related genes. Our qPCR results suggest that geranylgeranylation is the predominant form of prenylation in whole C. fumiferana.


Subject(s)
Farnesyltranstransferase/biosynthesis , Farnesyltranstransferase/genetics , Moths/enzymology , Amino Acid Sequence , Animals , Cloning, Molecular , Escherichia coli/genetics , Farnesyltranstransferase/chemistry , Kinetics , Ligands , Molecular Sequence Data , Moths/growth & development , Protein Prenylation/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sequence Analysis, DNA , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL