Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Eur J Biochem ; 268(18): 4850-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11559353

ABSTRACT

At least three different subcellular compartments, including peroxisomes, are involved in cholesterol biosynthesis. Because proper CNS development depends on de novo cholesterol biosynthesis, peroxisomes must play a critical functional role in this process. Surprisingly, no information is available on the peroxisomal isoprenoid/cholesterol biosynthesis pathway in normal brain tissue or on the compartmentalization of isoprene metabolism in the CNS. This has been due mainly to the lack of a well-defined isolation procedure for brain tissue, and also to the presence of myelin in brain tissue, which results in significant contamination of subcellular fractions. As a first step in characterizing the peroxisomal isoprenoid pathway in the CNS, we have established a purification procedure to isolate peroxisomes and other cellular organelles from the brain stem, cerebellum and spinal cord of the mouse brain. We demonstrate by use of marker enzymes and immunoblotting with antibodies against organelle specific proteins that the isolated peroxisomes are highly purified and well separated from the ER and mitochondria, and are free of myelin contamination. The isolated peroxisomal fraction was purified at least 40-fold over the original homogenate. In addition, we show by analytical subcellular fractionation and immunoelectron microscopy that HMG-CoA reductase protein and activity are localized both in the ER and peroxisomes in the CNS.


Subject(s)
Central Nervous System/cytology , Central Nervous System/enzymology , Hydroxymethylglutaryl CoA Reductases/analysis , Hydroxymethylglutaryl CoA Reductases/isolation & purification , Peroxisomes/enzymology , Phosphoric Diester Hydrolases , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase , 2',3'-Cyclic-Nucleotide Phosphodiesterases/metabolism , Animals , Brain Stem/cytology , Brain Stem/enzymology , Brain Stem/ultrastructure , Catalase/metabolism , Central Nervous System/ultrastructure , Centrifugation, Density Gradient , Cerebellum/cytology , Cerebellum/enzymology , Cerebellum/ultrastructure , Cholesterol/metabolism , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/ultrastructure , Hydroxymethylglutaryl CoA Reductases/ultrastructure , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent , Mice , Mice, Inbred ICR , Microscopy, Immunoelectron , Peroxisomes/ultrastructure , Spinal Cord/cytology , Spinal Cord/enzymology
2.
J Clin Invest ; 107(10): 1263-73, 2001 May.
Article in English | MEDLINE | ID: mdl-11375416

ABSTRACT

Hepatic steatosis is common in patients having severe hyperhomocysteinemia due to deficiency for cystathionine beta-synthase. However, the mechanism by which homocysteine promotes the development and progression of hepatic steatosis is unknown. We report here that homocysteine-induced endoplasmic reticulum (ER) stress activates both the unfolded protein response and the sterol regulatory element-binding proteins (SREBPs) in cultured human hepatocytes as well as vascular endothelial and aortic smooth muscle cells. Activation of the SREBPs is associated with increased expression of genes responsible for cholesterol/triglyceride biosynthesis and uptake and with intracellular accumulation of cholesterol. Homocysteine-induced gene expression was inhibited by overexpression of the ER chaperone, GRP78/BiP, thus demonstrating a direct role of ER stress in the activation of cholesterol/triglyceride biosynthesis. Consistent with these in vitro findings, cholesterol and triglycerides were significantly elevated in the livers, but not plasmas, of mice having diet-induced hyperhomocysteinemia. This effect was not due to impaired hepatic export of lipids because secretion of VLDL-triglyceride was increased in hyperhomocysteinemic mice. These findings suggest a mechanism by which homocysteine-induced ER stress causes dysregulation of the endogenous sterol response pathway, leading to increased hepatic biosynthesis and uptake of cholesterol and triglycerides. Furthermore, this mechanism likely explains the development and progression of hepatic steatosis and possibly atherosclerotic lesions observed in hyperhomocysteinemia.


Subject(s)
Cholesterol/metabolism , Endoplasmic Reticulum/metabolism , Heat-Shock Proteins , Homocysteine/metabolism , Hyperhomocysteinemia/metabolism , Liver/metabolism , Transcription Factors , Triglycerides/metabolism , Animals , Arteriosclerosis/etiology , CCAAT-Enhancer-Binding Proteins/metabolism , Carrier Proteins/metabolism , Cells, Cultured , Cystathionine beta-Synthase/genetics , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Chaperone BiP , Fatty Liver/etiology , Humans , Lipoproteins, VLDL/metabolism , Liver/cytology , Mice , Molecular Chaperones/metabolism , Protein Denaturation , Sterol Regulatory Element Binding Protein 1
3.
Biochim Biophys Acta ; 1529(1-3): 89-102, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11111079

ABSTRACT

At least three different subcellular compartments, including peroxisomes, are involved in cholesterol synthesis. Recently, it has been demonstrated that peroxisomes contain a number of enzymes involved in cholesterol biogenesis that previously were considered to be cytosolic or located in the endoplasmic reticulum. Peroxisomes have been shown to contain acetoacetyl-CoA thiolase, HMG-CoA synthase, HMG-CoA reductase, mevalonate kinase, phosphomevalonate kinase, phosphomevalonate decarboxylase, isopentenyl diphosphate isomerase and FPP synthase. Moreover, the activities of these enzymes are also significantly decreased in liver tissue and fibroblast cells obtained from patients with peroxisomal deficiency diseases. In addition, the cholesterol biosynthetic capacity is severely impaired in cultured skin fibroblasts obtained from patients with peroxisomal deficiency diseases. These findings support the proposal that peroxisomes play an essential role in isoprenoid biosynthesis. This paper presents a review of peroxisomal protein targeting and of recent studies demonstrating the localization of cholesterol biosynthetic enzymes in peroxisomes and the identification of peroxisomal targeting signals in these proteins.


Subject(s)
Cholesterol/biosynthesis , Peroxisomes/enzymology , Proteins/metabolism , Amino Acid Sequence , Animals , Biological Transport , Cell Line , Cholesterol/metabolism , Cytosol/metabolism , Fibroblasts/metabolism , Humans , Membrane Proteins/metabolism , Mitochondria/metabolism , Peroxisomal Disorders/enzymology , Signal Transduction , Transfection
4.
J Lipid Res ; 41(12): 1921-35, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11108725

ABSTRACT

At least three different subcellular compartments, including peroxisomes, are involved in cholesterol synthesis. The peroxisomal targeting signals for phosphomevalonate kinase and isopentenyl diphosphate isomerase have been identified. In the current study we identify the peroxisomal targeting signals required for four other enzymes of the cholesterol biosynthetic pathway: acetoacetyl-CoA (AA-CoA) thiolase, 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) synthase, mevalonate diphosphate decarboxylase (MPPD), and farnesyl diphosphate (FPP) synthase. Data are presented that demonstrate that mitochondrial AA-CoA thiolase contains both a mitochondrial targeting signal at the amino terminus and a peroxisomal targeting signal (PTS-1) at the carboxy terminus. We also analyze a new variation of PTS-2 sequences required to target HMG-CoA synthase and MPPD to peroxisomes. In addition, we show that FPP synthase import into peroxisomes is dependent on the PTS-2 receptor and identify at the amino terminus of the protein a 20-amino acid region that is required for the peroxisomal localization of the enzyme. These data provide further support for the conclusion that peroxisomes play a critical role in cholesterol biosynthesis.


Subject(s)
Acetyl-CoA C-Acetyltransferase/metabolism , Alkyl and Aryl Transferases/metabolism , Carboxy-Lyases/metabolism , Hydroxymethylglutaryl-CoA Synthase/metabolism , Peroxisomes/metabolism , Alkyl and Aryl Transferases/chemistry , Amino Acid Sequence , Animals , Base Sequence , CHO Cells , Cricetinae , DNA Primers , Geranyltranstransferase , Microscopy, Immunoelectron , Mitochondria/enzymology , Molecular Sequence Data , Peroxisomes/ultrastructure
5.
J Lipid Res ; 41(8): 1347-55, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10946023

ABSTRACT

CHO cells expressing the liver-specific gene product cholesterol-7alpha-hydroxylase showed a 6-fold increase in the biosynthesis of [(14)C]cholesterol from [(14)C]acetate, as well as increased enzymatic activities of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase and squalene synthase. Cells expressing cholesterol-7alpha-hydroxylase contained less sterol response element-binding protein 1 (SREBP1) precursor, whereas the cellular content of mature SREBP1, as well as the mRNAs of cholesterol biosynthetic genes (HMG-CoA reductase and squalene synthase), were all increased approximately 3-fold. Cells expressing cholesterol-7alpha-hydroxylase displayed greater activities of luciferase reporters containing the SREBP-dependent promoter elements derived from HMG-CoA reductase and farnesyl diphosphate synthase, in spite of accumulating significantly more free and esterified cholesterol and 7alpha-hydroxycholesterol. While cells expressing cholesterol-7alpha-hydroxylase displayed increased SREBP-dependent transcription, sterol-mediated repression of SREBP-dependent transcription by LDL-cholesterol and exogenous oxysterols was similar in both cell types. Cells expressing cholesterol-7alpha-hydroxylase displayed greater rates of secretion of cholesterol as well as increased expression of the ABC1 cassette protein mRNA. Adding 25-hydroxycholesterol to the culture medium of both cell types increased the expression of ABC1 cassette protein mRNA. The combined data suggest that in nonhepatic CHO cells multiple regulatory processes sensitive to cellular sterols act independently to coordinately maintain cellular cholesterol homeostasis.


Subject(s)
CCAAT-Enhancer-Binding Proteins , CHO Cells/enzymology , Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol/biosynthesis , Cholesterol/metabolism , Gene Expression , Homeostasis , Transcription Factors , Acetates/metabolism , Animals , Cells, Cultured , Cholesterol 7-alpha-Hydroxylase/metabolism , Cholesterol Esters/metabolism , Cricetinae , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/pharmacology , Farnesyl-Diphosphate Farnesyltransferase/genetics , Farnesyl-Diphosphate Farnesyltransferase/metabolism , Hydroxycholesterols/pharmacology , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl CoA Reductases/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/pharmacology , Promoter Regions, Genetic , RNA, Messenger/metabolism , Rats , Response Elements , Sterol Regulatory Element Binding Protein 1 , Transcription, Genetic/drug effects
6.
Biochemistry ; 39(1): 237-47, 2000 Jan 11.
Article in English | MEDLINE | ID: mdl-10625499

ABSTRACT

We have previously identified a CHO cell line (UT2 cells) that expresses only one 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase protein which is localized exclusively in peroxisomes [Engfelt, H.W., Shackelford, J.E., Aboushadi, N., Jessani, N., Masuda, K., Paton, V.G., Keller, G.A., and Krisans, S.K. (1997) J. Biol. Chem. 272, 24579-24587]. In this study, we utilized the UT2 cells to determine the properties of the peroxisomal reductase independent of the endoplasmic reticulum (ER) HMG-CoA reductase. We demonstrated major differences between the two proteins. The peroxisomal reductase is not the rate-limiting enzyme for cholesterol biosynthesis in UT2 cells. The peroxisomal reductase protein is not phosphorylated, and its activity is not altered in the presence of inhibitors of cellular phosphatases. Its rate of degradation is not accelerated in response to mevalonate. Finally, the degradation process is not blocked by N-acetyl-Leu-Leu-norleucinal (ALLN). Furthermore, the peroxisomal HMG-CoA reductase is significantly more resistant to inhibition by statins. Taken together, the data support the conclusion that the peroxisomal reductase is functionally and structurally different from the ER HMG-CoA reductase.


Subject(s)
Cholesterol/biosynthesis , Hydroxymethylglutaryl CoA Reductases/chemistry , Hydroxymethylglutaryl CoA Reductases/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/pharmacology , Peroxisomes/enzymology , Acetic Acid/metabolism , Acetyl-CoA C-Acetyltransferase/metabolism , Adenosine Triphosphate/metabolism , Animals , CHO Cells/drug effects , CHO Cells/enzymology , Carbon Radioisotopes , Cell Cycle , Cell Survival , Clone Cells/drug effects , Clone Cells/enzymology , Cricetinae , Deuterium Oxide/metabolism , Enzyme Activation/drug effects , Fatty Acids, Unsaturated/pharmacology , Hydroxymethylglutaryl CoA Reductases/biosynthesis , Hydroxymethylglutaryl-CoA Synthase/metabolism , Lactones/pharmacology , Leupeptins/pharmacology , Mevalonic Acid/metabolism , Phosphorylation , Simvastatin/pharmacology , Tritium
7.
J Histochem Cytochem ; 47(9): 1127-32, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10449533

ABSTRACT

Our group and others have recently demonstrated that peroxisomes contain a number of enzymes involved in cholesterol biosynthesis that previously were considered to be cytosolic or located in the endoplasmic reticulum (ER). Peroxisomes have been shown to contain HMG-CoA reductase, mevalonate kinase, phosphomevalonate kinase, phosphomevalonate decarboxylase, isopentenyl diphosphate isomerase, and FPP synthase. Four of the five enzymes required for the conversion of mevalonate to FPP contain a conserved putative PTS1 or PTS2, supporting the concept of targeted transport into peroxisomes. To date, no information is available regarding the function of the peroxisomal HMG-CoA reductase in cholesterol/isoprenoid metabolism, and the structure of the peroxisomal HMG-CoA reductase has yet to be determined. We have identified a mammalian cell line that expresses only one HMG-CoA reductase protein, and which is localized exclusively to peroxisomes, to facilitate our studies on the function, regulation, and structure of the peroxisomal HMG-CoA reductase. This cell line was obtained by growing UT2 cells (which lack the ER HMG-CoA reductase) in the absence of mevalonate. The surviving cells exhibited a marked increase in a 90-kD HMG-CoA reductase that was localized exclusively to peroxisomes. The wild-type CHO cells contain two HMG-CoA reductase proteins, the well-characterized 97-kD protein localized in the ER, and a 90-kD protein localized in peroxisomes. We have also identified the mutations in the UT2 cells responsible for the lack of the 97-kD protein. In addition, peroxisomal-deficient Pex2 CHO cell mutants display reduced HMG-CoA reductase levels and have reduced rates of sterol and nonsterol biosynthesis. These data further support the proposal that peroxisomes play an essential role in isoprenoid biosynthesis.


Subject(s)
Microbodies/enzymology , Protein Prenylation , Acyl Coenzyme A/genetics , Acyl Coenzyme A/metabolism , Animals , CHO Cells , Carbon-Carbon Double Bond Isomerases/genetics , Carbon-Carbon Double Bond Isomerases/metabolism , Cells, Cultured , Cholesterol/metabolism , Cricetinae , Hemiterpenes , Humans , Microscopy, Fluorescence , Mutation , Rats , Transfection
8.
Am J Hum Genet ; 65(2): 327-35, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10417275

ABSTRACT

Mevalonate kinase (MKase) deficiency (MKD) is a rare autosomal recessive disorder in the pathway of cholesterol and nonsterol isoprenoid biosynthesis. Thus far, two disease-causing missense alleles have been identified, N301T and A334T. We report four additional mutations associated with MKD: L264F, T243I, L265P, and I268T, the last found in a patient of Mennonite ancestry. Electrophoretic analysis of bacterially expressed wild-type and mutant MKase indicated that I268T and T243I mutants produced normal or somewhat reduced amounts of MKase protein; conversely, L264F and L265P mutations resulted in considerably decreased, or absent, MKase protein. Immunoblot analysis of MKase from all patients suggested that the MKase polypeptide was grossly intact and produced in amounts comparable to control levels. Three mutations resulted in significantly diminished MKase enzyme activity (<2%), whereas the I268T allele yielded approximately 20% residual enzyme activity. Our results should allow more-accurate identification of carriers and indicate a mutation "cluster" within amino acids 240-270 of the mature MKase polypeptide.


Subject(s)
Metabolism, Inborn Errors/genetics , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/genetics , Alleles , Amino Acid Sequence , Amino Acid Substitution , Cells, Cultured , Christianity , DNA Mutational Analysis , Escherichia coli/genetics , Female , Fibroblasts/enzymology , Fibroblasts/metabolism , Humans , Lymphocytes/enzymology , Lymphocytes/metabolism , Male , Metabolism, Inborn Errors/enzymology , Molecular Sequence Data , Mutation/genetics , Nuclear Family , Pedigree , Phosphotransferases (Alcohol Group Acceptor)/isolation & purification , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Sequence Alignment
9.
J Lipid Res ; 40(4): 672-9, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10191291

ABSTRACT

Phosphomevalonate kinase catalyzes the conversion of mevalonate-5-phosphate to mevalonate-5-diphosphate and was originally believed to be a cytosolic enzyme. In this study we have localized the phosphomevalonate kinase gene to chromosome 1p13-1q22-23 and present a genomic map indicating that the gene spans more than 8.4 kb in the human genome. Furthermore, we show that message levels and enzyme activity of rat liver phosphomevalonate kinase are regulated in response to dietary sterol levels and that this regulation is coordinate with 3-hydroxy-3-methylglutaryl coenzyme A reductase, the rate-limiting enzyme of cholesterol biosynthesis. In addition, we demonstrate that phosphomevalonate kinase is a peroxisomal protein which requires the C-terminal peroxisomal targeting signal, Ser-Arg-Leu, for localization to the organelle.


Subject(s)
Chromosome Mapping , Gene Expression Regulation, Enzymologic , Liver/ultrastructure , Phosphotransferases (Phosphate Group Acceptor)/genetics , Amino Acid Sequence , Animals , Chromosomes, Human, Pair 1 , Gene Expression Regulation, Enzymologic/drug effects , Gene Targeting , Humans , Liver/enzymology , Male , Microbodies/enzymology , Molecular Sequence Data , Phosphotransferases (Phosphate Group Acceptor)/chemistry , Phosphotransferases (Phosphate Group Acceptor)/metabolism , Protein Sorting Signals/chemistry , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Sequence Alignment , Sterols/administration & dosage
10.
J Lipid Res ; 39(11): 2182-91, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9799804

ABSTRACT

UT2 cells are a mutant clone of Chinese hamster ovary (CHO) cells that are deficient in the 97 kDa endoplasmic reticulum (ER) 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase protein. The analysis of UT2 cell cDNA and genomic DNA has led to the identification of two novel point mutations in intronic sequences of the ER HMG-CoA reductase gene. One mutation identified at the +1 position (G --> A) of the 5' splice site of exon 11-12 junction was shown to cause exon 11 skipping which resulted in the insertion of premature stop codons. We also identified a second mutation at the +5 position (G --> A) of the 5' splice site in the intron spanning exons 13 and 14. Furthermore, the data indicate that the two mutations in the reductase gene are present on the same allele. As demonstrated by reverse transcription-polymerase chain reaction (RT-PCR) of UT2 cell mRNA, the mutations produce aberrant spliced messages. If the aberrant messages were translated, truncated proteins of 44 kDa or 66 kDa would be predicted. More importantly, these truncated proteins would be expected not to have catalytic activity. In addition, we have also recently demonstrated that the UT2 cells express a 90 kDa HMG-CoA reductase protein that is localized exclusively in peroxisomes, and is up-regulated when the cells are grown in the absence of added mevalonate. Thus, the mutations identified in the ER reductase gene in UT2 cells indicate that neither a 97 kDa nor a 90 kDa reductase protein can be produced from this gene.


Subject(s)
Endoplasmic Reticulum/enzymology , Hydroxymethylglutaryl CoA Reductases/genetics , Mutation , RNA Splicing/genetics , Alleles , Animals , Blotting, Northern , Blotting, Southern , CHO Cells , Cricetinae , DNA, Complementary/chemistry , Exons , Molecular Weight , Polymorphism, Restriction Fragment Length , Sequence Analysis, DNA
11.
J Lipid Res ; 39(9): 1781-91, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9741690

ABSTRACT

ZR-78 and ZR-82 cells are two peroxisomal-deficient Chinese hamster ovary (CHO) cell mutants. These cells lack normal peroxisomes and show reduced levels of plasmalogen synthesis and other peroxisomal functions attributed to the deficiency of peroxisomal matrix enzymes. As we have recently identified two HMG-CoA reductase proteins in CHO cells, a 97 kDa reductase localized in the ER and a 90 kDa reductase protein localized in peroxisomes, this enabled us to study the two reductase proteins for the first time in peroxisomal-deficient CHO cells. In this study we report the results of a detailed analysis of the isoprenoid biosynthetic pathway in the peroxisomal-deficient CHO cell lines ZR-78 and ZR-82. We demonstrate that total HMG-CoA reductase activity is significantly reduced in the peroxisomal-deficient cells as compared to the wild type cells. Analysis of the two reductase proteins in permeabilized cells indicated that in the ZR-78 and ZR-82 cells the 90 kDa peroxisomal reductase protein was mainly localized to the cytosol. We further show that the rates of both sterol (cholesterol) and non-sterol (dolichols) biosynthesis were significantly lower in the peroxisomal-deficient cells, when either [3H] acetate or [3H] mevalonate was used as substrate. In contrast, the rate of dolichol biosynthesis in the peroxisomal-deficient cells was similar to that of the wild type cells when incubated with [3H] farnesol. In addition, we demonstrate that the peroxisomal-deficient cells exhibited increased rates of lanosterol biosynthesis as compared to wild type cells. The results of this study provide further evidence for the essential requirement of peroxisomes for cholesterol biosynthesis as well as for dolichol production.


Subject(s)
Microbodies/physiology , Polyisoprenyl Phosphates/biosynthesis , Acetates/metabolism , Animals , CHO Cells , Cell Membrane Permeability , Cholesterol/biosynthesis , Cricetinae , Cytosol/metabolism , Dolichol Phosphates/biosynthesis , Farnesol/metabolism , Hydroxymethylglutaryl CoA Reductases/metabolism , Immunoblotting , Lanosterol/biosynthesis , Mevalonic Acid/metabolism , Microbodies/enzymology , Mutation , Tritium
12.
J Biol Chem ; 272(42): 26756-60, 1997 Oct 17.
Article in English | MEDLINE | ID: mdl-9334262

ABSTRACT

Sequencing of polymerase chain reaction-amplified cDNAs from cultured cells of three patients with mevalonate kinase deficiency revealed a G --> A transversion at nucleotide 1000 of the coding region, converting alanine to threonine at position 334 (A334T). To characterize this defect, we expressed wild-type and mutant cDNAs in Escherichia coli as the glutathione S-transferase fusion proteins, with purification by affinity chromatography. SDS-polyacrylamide gel electrophoresis analysis for wild-type and mutant fusion proteins indicated an expected molecular mass of 42-43 kDa. Kinetic characterization of the wild-type fusion protein yielded Km values of 150 +/- 23 and 440 +/- 190 microM (mean +/- S.E.) for substrates (RS)-mevalonate and ATP, respectively. Expressed wild-type mevalonate kinase (MKase) had a maximum velocity of 13.6 +/- 1.4 units/mg of protein (n = 22, +/-S.E.), whereas the A334T mutation yielded an enzyme with average Vmax of 0.26 +/- 0.02 unit/mg of protein (n = 6, +/-S.E.), representing a decrease to 1.4% of control Vmax. Restriction digestion with HhaI, in conjunction with direct sequencing of cDNAs, revealed that two patients were homozygous and one heterozygous for the A334T allele, establishing autosomal recessive inheritance within families. Although the A334T enzyme had a normal Km for ATP of 680 +/- 226 microM (n = 3, +/-S.E.), the Michaelis constant for (RS)-mevalonate was increased >30-fold to 4623 +/- 1167 microM (n = 4, +/-S.E.) under standard assay conditions. Comparable kinetic results were obtained using extracts of lymphoblasts, which were homozygous for the A334T allele. Alanine 334 is invariant in MKase from bacteria to man and located in a glycine-rich region postulated to have homology with ATP-binding sequences. Our results indicate that the bacterial expression system for human MKase will provide a useful model system in which to analyze inherited mutations and identify the first active site residue in MKase associated with stabilization of mevalonate binding.


Subject(s)
Alanine/metabolism , Metabolism, Inborn Errors/genetics , Mutation , Phosphotransferases (Alcohol Group Acceptor)/genetics , Alanine/genetics , Alleles , Binding Sites , Homozygote , Humans , Kinetics , Lymphocytes/enzymology , Mevalonic Acid/metabolism , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/metabolism
13.
J Biol Chem ; 272(39): 24579-87, 1997 Sep 26.
Article in English | MEDLINE | ID: mdl-9305924

ABSTRACT

In the liver 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase is present not only in the endoplasmic reticulum but also in the peroxisomes. However, to date no information is available regarding the function of the peroxisomal HMG-CoA reductase in cholesterol/isoprenoid metabolism, and the structure of the peroxisomal HMG-CoA reductase has yet to be determined. We have identified a mammalian cell line that expresses only one HMG-CoA reductase protein and that is localized exclusively to peroxisomes. This cell line was obtained by growing UT2 cells (which lack the endoplasmic reticulum HMG-CoA reductase) in the absence of mevalonate. The cells exhibited a marked increase in a 90-kDa HMG-CoA reductase that was localized exclusively to peroxisomes. The wild type Chinese hamster ovary cells contain two HMG-CoA reductase proteins, the well characterized 97-kDa protein, localized in the endoplasmic reticulum, and a 90-kDa protein localized in peroxisomes. The UT2 cells grown in the absence of mevalonate containing the up-regulated peroxisomal HMG-CoA reductase are designated UT2*. A detailed characterization and analysis of this cell line is presented in this study.


Subject(s)
Hydroxymethylglutaryl CoA Reductases/biosynthesis , Microbodies/enzymology , Animals , Blotting, Western , CHO Cells , Cell Extracts , Cell Fractionation , Cell Line , Centrifugation , Clone Cells , Cricetinae , Enzyme Induction , Hydroxymethylglutaryl CoA Reductases/immunology , Liver/enzymology , Male , Microscopy, Fluorescence , Rats , Rats, Sprague-Dawley
14.
J Biol Chem ; 272(30): 18945-50, 1997 Jul 25.
Article in English | MEDLINE | ID: mdl-9228075

ABSTRACT

To date, isopentenyl diphosphate:dimethylallyl diphosphate isomerase (IPP isomerase; EC 5.3.3.2) is presumed to have a cytosolic localization. However, we have recently shown that in permeabilized cells lacking cytosolic components, mevalonate can be converted to cholesterol, implying that all of the enzymes required for the conversion of mevalonate to farnesyl diphosphate are found in the peroxisome. To provide unequivocal evidence for the subcellular localization of IPP isomerase, in this study, we have cloned the rat and hamster homologues of IPP isomerase and identified the signal that targets this enzyme to peroxisomes. In addition, we also demonstrate that IPP isomerase is regulated at the mRNA level.


Subject(s)
Carbon-Carbon Double Bond Isomerases , Isomerases/metabolism , Microbodies/enzymology , Amino Acid Sequence , Animals , Base Sequence , Biological Transport , Blotting, Southern , Cells, Cultured , Cholesterol/biosynthesis , Cloning, Molecular , Consensus Sequence , Cricetinae , Dietary Proteins/metabolism , Hemiterpenes , Human Genome Project , Humans , Isomerases/genetics , Liver/cytology , Liver/enzymology , Male , Mevalonic Acid/metabolism , Molecular Sequence Data , Peroxisome-Targeting Signal 1 Receptor , Protein Sorting Signals/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/metabolism
15.
Biochem Biophys Res Commun ; 230(3): 562-8, 1997 Jan 23.
Article in English | MEDLINE | ID: mdl-9015362

ABSTRACT

In this study we provide evidence for the first time that rat liver microsomal and peroxisomal fractions are able to phosphorylate free farnesol to its diphosphate ester in a CTP dependent manner. The farnesyl diphosphate (FPP) kinase activity is decreased in whole liver homogenates obtained from rats treated with cholesterol and unchanged in homogenates obtained from rats treated with cholestyramine. In contrast, farnesyl pyrophosphatase (FPPase) activity, an enzyme which specifically hydrolyzes FPP to farnesol is only found in the microsomal fraction and is unaffected by treatment of rats with cholesterol or cholestyramine. In addition, we also demonstrate that farnesol can be oxidized to a prenyl aldehyde, presumably by an alcohol dehydrogenase (ADH), and that this activity resides in the mitochondrial and peroxisomal fractions.


Subject(s)
Farnesol/metabolism , Microbodies/enzymology , Microbodies/metabolism , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Alcohol Dehydrogenase/metabolism , Animals , Cell Fractionation , Cholesterol/biosynthesis , Cholesterol, Dietary/pharmacology , Male , Phosphorylation , Polyisoprenyl Phosphates/metabolism , Pyrophosphatases/metabolism , Rats , Sesquiterpenes , Substrate Specificity
16.
Ann N Y Acad Sci ; 804: 142-64, 1996 Dec 27.
Article in English | MEDLINE | ID: mdl-8993542

ABSTRACT

Thus, the results showing the presence of cholesterol synthetic enzymes in peroxisomes (see references 1, 4, 5, 6, 7, 8, 12, 13, 20, 21, 22, 24, 25, and 26), the reduced levels of cholesterol synthesis enzymes and cholesterol synthetic capacity of cells and tissues lacking peroxisomes, 26, 37, 39 and the low serum cholesterol levels in patients suffering from peroxisomal deficiency diseases40-43 demonstrate that peroxisomes are essential for normal cholesterol synthesis. A number of metabolic pathways require co-participation of enzymes located in both peroxisomes as well as enzymes found in other intracellular compartments. For example, the first steps of plasmalogen synthesis occur in the peroxisomes, while the terminal reactions are completed in the endoplasmic reticulum. Similarly, the oxidation of cholesterol to bile acids requires the participation of enzymes localized in the endoplasmic reticulum as well as peroxisomes. Little is known about the regulation of such pathways or about the shuttling of intermediates between compartments. The physiological importance of peroxisomal enzymes in the regulation of sterol metabolism remains to be clarified.


Subject(s)
Cholesterol/biosynthesis , Microbodies/metabolism , Plant Proteins , Acetyl-CoA C-Acyltransferase/metabolism , Animals , Carboxy-Lyases/metabolism , Carrier Proteins/metabolism , Cell Compartmentation , Farnesyl-Diphosphate Farnesyltransferase/metabolism , Humans , Hydroxymethylglutaryl CoA Reductases/metabolism , Hydroxymethylglutaryl-CoA Synthase/metabolism , Lanosterol/analogs & derivatives , Lanosterol/metabolism , Liver/enzymology , Mevalonic Acid/metabolism , Peroxisomal Disorders/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Phosphate Group Acceptor)/metabolism , Polyisoprenyl Phosphates/metabolism , Rats , Sesquiterpenes , Squalene/metabolism
17.
J Biol Chem ; 271(3): 1784-8, 1996 Jan 19.
Article in English | MEDLINE | ID: mdl-8576183

ABSTRACT

We have recently demonstrated that mevalonate kinase and farnesyl diphosphate (FPP) synthase are localized predominantly in peroxisomes. This observation raises the question regarding the subcellular localization of the enzymes that catalyze the individual steps in the pathway between mevalonate kinase and FPP synthase (phosphomevalonate kinase, mevalonate diphosphate decarboxylase, and isopentenyl diphosphate isomerase). These enzyme are found in the 100,000 x g supernatant fraction of cells or tissues and have been considered to be cytoplasmic proteins. In the current studies, we show that the activities of mevalonate kinase, phosphomevalonate kinase, and mevalonate diphosphate decarboxylase are equal in extracts prepared from intact cells and selectively permeabilized cells, which lack cytosolic enzymes. We also demonstrate structure-linked latency of phosphomevalonate kinase and mevalonate diphosphate decarboxylase that is consistent with a peroxisomal localization of these enzymes. Finally, we show that cholesterol biosynthesis from mevalonate can occur in selectively permeabilized cells lacking cytosolic components. These results suggest that the peroxisome is the major site of the synthesis of FPP from mevalonate, since all of the cholestrogenic enzymes involved in this conversion are localized in the peroxisome.


Subject(s)
Alkyl and Aryl Transferases , Carbon-Carbon Double Bond Isomerases , Cholesterol/biosynthesis , Mevalonic Acid/metabolism , Microbodies/metabolism , Polyisoprenyl Phosphates/metabolism , Animals , Carboxy-Lyases/analysis , Carboxy-Lyases/metabolism , Cell Line , Cell Membrane Permeability , Chlorocebus aethiops , Cytosol/enzymology , Geranyltranstransferase , Hemiterpenes , Isomerases/analysis , Isomerases/metabolism , Kidney , Kinetics , Phosphotransferases (Alcohol Group Acceptor)/analysis , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Sesquiterpenes , Subcellular Fractions/enzymology , Transferases/analysis , Transferases/metabolism
18.
Genomics ; 24(1): 14-9, 1994 Nov 01.
Article in English | MEDLINE | ID: mdl-7896269

ABSTRACT

Human cellular nucleic acid binding protein (CNBP) is a zinc finger DNA binding protein of unknown function. The human CNBP cDNA was used as a probe to isolate four structurally distinct but highly homologous mouse liver cDNA clones. Each of the mouse clones exhibited extraordinary sequence conservation with human CNBP cDNA, and the predicted mouse amino acid sequence identities with human CNBP protein ranged from 99 to 100%. Genetic mapping of CNBP genes in interspecific and intersubspecific mouse backcrosses revealed two loci that hybridize to CNBP cDNA at high stringency, located on chromosomes 5 and 6. The subcellular distribution of the CNBP protein was characterized with a specific polyclonal antibody generated against a synthetic peptide from the carboxyl terminus. CNBP was found in the cytosol and the endoplasmic reticulum in subcellular fractions from mouse liver, but was undetectable in nuclear fractions. These data suggest that CNBP is a member of a highly conserved family of cytosolic proteins that may be encoded by multiple dispersed genes.


Subject(s)
DNA-Binding Proteins/genetics , RNA-Binding Proteins , Zinc Fingers , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , DNA, Complementary , DNA-Binding Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Molecular Sequence Data , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Subcellular Fractions/metabolism
19.
J Biol Chem ; 269(19): 14165-9, 1994 May 13.
Article in English | MEDLINE | ID: mdl-8188698

ABSTRACT

In this study, we have investigated the subcellular localization of farnesyl-diphosphate synthase (FPP synthase). FPP synthase produces FPP, which is utilized in the synthesis of squalene, cholesterol, farnesylated and geranylgeranylated proteins, dolichols, coenzyme Q, and the isoprenoid moiety of heme a. This enzyme is found in the 100,000 x g supernatant fraction of cells or tissues and has been considered to be a cytoplasmic protein. In this study, analysis of FPP synthase activity and protein in fractionated rat liver together with immunofluorescent and immunoelectron microscopy studies demonstrated unequivocally that FPP synthase is largely localized in peroxisomes. These data, in combination with the previous observation that mevalonate kinase is predominantly localized in peroxisomes, suggest that peroxisomes are the major site of synthesis of FPP from mevalonate. We also demonstrate that in liver tissue obtained from patients with peroxisomal deficiency diseases (Zellweger syndrome and neonatal adrenoleukodystrophy), the activities of five enzymes involved in isoprenoid synthesis, namely mevalonate kinase, phosphomevalonate kinase, mevalonate-diphosphate decarboxylase, isopentenyl-diphosphate isomerase, and FPP synthase, are significantly reduced, consistent with a peroxisomal localization of these enzymes.


Subject(s)
Alkyl and Aryl Transferases , Microbodies/enzymology , Transferases/metabolism , Adrenoleukodystrophy/enzymology , Animals , Centrifugation, Density Gradient , Geranyltranstransferase , Humans , Liver/enzymology , Liver/ultrastructure , Male , Microbodies/ultrastructure , Microscopy, Fluorescence , Microscopy, Immunoelectron , Rats , Rats, Sprague-Dawley , Zellweger Syndrome/enzymology
20.
J Biol Chem ; 269(2): 1197-205, 1994 Jan 14.
Article in English | MEDLINE | ID: mdl-7904598

ABSTRACT

We reported recently that mevalonate kinase (EC 2.7.1.36; ATP:mevalonate 5-phosphotransferase) that was isolated from rat liver and believed to be a cytosolic protein was localized in rat liver peroxisomes. In addition, we found that the mevalonate kinase monoclonal antibody used in the study also reacted with several other proteins present in the mitochondrial and cytosolic fractions. These findings raised the prospect of the presence of several isoenzymes of mevalonate kinase localized in different compartments of the cell. In the current study we produced four new polyclonal antibodies against different epitopes of mevalonate kinase to investigate the subcellular localization of the protein by several different approaches: (i) by analytical subcellular fractionation and immunoblotting of mevalonate kinase in the isolated subcellular fractions with the monospecific antibodies; (ii) by immunocryoelectron microscopy techniques; and (iii) by expressing the cDNA encoding mevalonate kinase in mammalian cells. The data obtained demonstrate that there is only one mevalonate kinase protein that is predominantly localized in peroxisomes. We also illustrate that the protein is targeted to and imported into peroxisomes. In addition, we show that in cells and tissues obtained from patients with peroxisomal deficiency diseases mevalonate kinase protein and its activity are severely reduced.


Subject(s)
Adrenoleukodystrophy/enzymology , Microbodies/enzymology , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Zellweger Syndrome/enzymology , Acetyl-CoA C-Acetyltransferase/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Cell Compartmentation , Cloning, Molecular , Liver/enzymology , Male , Microscopy, Electron , Molecular Sequence Data , Molecular Weight , Phosphotransferases (Alcohol Group Acceptor)/immunology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...