Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Gene ; 278(1-2): 185-92, 2001 Oct 31.
Article in English | MEDLINE | ID: mdl-11707336

ABSTRACT

Acyl-CoA synthetases (ACSs) play an essential role in fatty acid metabolism. ACS3 is an arachidonate-preferring enzyme expressed in a wide range of human tissues including brain, heart, placenta, prostate, skeletal muscle, testis and thymus. As an initial step to understanding the transcriptional regulation of the human ACS3 gene, we analyzed the genomic organization and transcription units of the human ACS3 gene. Sequence analysis of genomic clones demonstrates that the human ACS3 gene spans at least 80.6 kb and contains 17 exons. The human ACS3 gene was mapped between the sequence-tagged site markers D2S360 and WI-21901. Sequence inspection of the 5'-flanking region revealed potential DNA elements including CCAAT, AP-1, Oct-1, GATAs, SRY, CdxA, Nkx-2.5, c-Myb, HSF2, NF-AT, AP-2, NF-Y, and p300. A minimal promoter region required for the expression of the human ACS3 gene in melanoma G361 cells was determined.


Subject(s)
Coenzyme A Ligases/genetics , Genes/genetics , Repressor Proteins , Saccharomyces cerevisiae Proteins , Transcription, Genetic/genetics , 5' Flanking Region/genetics , Amino Acid Sequence , Base Sequence , Chromosome Mapping , Chromosomes, Human, Pair 2/genetics , DNA/chemistry , DNA/genetics , DNA/isolation & purification , Exons , Female , Gene Expression , HL-60 Cells , HeLa Cells , Humans , Introns , K562 Cells , Luciferases/genetics , Luciferases/metabolism , Male , Molecular Sequence Data , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Tissue Distribution , Tumor Cells, Cultured
2.
Biochem Biophys Res Commun ; 286(1): 80-6, 2001 Aug 10.
Article in English | MEDLINE | ID: mdl-11485311

ABSTRACT

Acyl-CoA synthetase 4 (ACS4) is an arachidonate-preferring isozyme of ACS family predominantly expressed in steroidogenic tissues. Isolation and characterization of genomic clones encoding human ACS4 revealed that the genomic organization of the gene. The human ACS4 gene spans approximately 90 kb and consists of 16 exons. Sequence inspection of the 5'-flanking region revealed potential DNA elements including GATAs, p300, AP-4, SRY, CREB and MyoD. A minimal promoter region required for the expression of ACS4 in HeLa S3 cells was determined. The human ACS4 gene was mapped between the STS markers, WI-17685 and CHLC.GATA81B07 on Xq22-23 region.


Subject(s)
Coenzyme A Ligases/genetics , Exons , Introns , Transcription, Genetic , Amino Acid Sequence , Base Sequence , DNA , HeLa Cells , Humans , Molecular Sequence Data , Sequence Homology, Amino Acid
3.
J Biol Chem ; 276(36): 34259-69, 2001 Sep 07.
Article in English | MEDLINE | ID: mdl-11435428

ABSTRACT

Cytosolic acetyl-CoA synthetase (AceCS1) activates acetate to supply the cells with acetyl-CoA for lipid synthesis. The cDNA for the mammalian AceCS1 has been isolated recently, and the mRNA was shown to be negatively regulated by sterols in cultured cells. In the current study, we describe the molecular mechanisms directing the sterol-regulated expression of murine AceCS1 by cloning and functional studies of the 5'-flanking region of the AceCS1 gene. An AceCS1 promoter-reporter gene (approximately 2.1 kilobase pairs) was negatively regulated when sterols were added to the medium of cultured cells, and the promoter was markedly induced by co-transfection of a plasmid that expresses the transcriptionally active nuclear form of either sterol regulatory element-binding protein (SREBP)-1a or -2 in HepG2 cells. Sequence analysis suggested that the AceCS1 promoter contains an E-box, two putative CCAAT-boxes, eight sterol regulatory element (SRE) motifs, and six GC-boxes. Gel shift assays demonstrated that all eight SRE motifs bound purified SREBP-1a in vitro with similar affinity. Luciferase reporter gene assays revealed that sterol regulation was critically dependent on three closely spaced SRE motifs and an adjacent GC-box. However, mutation of two putative upstream CCAAT-boxes did not affect SREBP dependent activation. Electrophoretic mobility "supershift" analyses confirmed that both Sp1 and Sp3 bound to the critical GC-box. In addition, transfection studies in Drosophila SL2 cells demonstrated that SREBP synergistically activated the AceCS1 promoter along with Sp1 or Sp3 but not with nuclear factor-Y.


Subject(s)
Acetate-CoA Ligase/genetics , Gene Expression Regulation, Enzymologic , Transcription Factors , Transcription, Genetic , Amino Acid Motifs , Animals , Base Sequence , Binding Sites , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line , Cell Nucleus/metabolism , Cells, Cultured , DNA Mutational Analysis , DNA, Complementary/metabolism , DNA-Binding Proteins/metabolism , Drosophila , Gene Deletion , Genes, Reporter , Humans , Luciferases/metabolism , Mice , Molecular Sequence Data , Multigene Family , Mutation , Plasmids/metabolism , Promoter Regions, Genetic , Protein Isoforms , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Sp1 Transcription Factor/metabolism , Sterol Regulatory Element Binding Protein 1 , Transcriptional Activation , Transfection
4.
J Biol Chem ; 276(38): 35961-6, 2001 Sep 21.
Article in English | MEDLINE | ID: mdl-11470804

ABSTRACT

In this study, we identified and characterized two murine cDNAs encoding medium-chain acyl-CoA synthetase (MACS). One, designated MACS1, is a novel protein and the other the product of the Sa gene (Sa protein), which is preferentially expressed in spontaneously hypertensive rats. Based on the murine MACS1 sequence, we also identified the location and organization of the human MACS1 gene, showing that the human MACS1 and Sa genes are located in the opposite transcriptional direction within a 150-kilobase region on chromosome 16p13.1. Murine MACS1 and Sa protein were overexpressed in COS cells, purified to homogeneity, and characterized. Among C4-C16 fatty acids, MACS1 preferentially utilizes octanoate, whereas isobutyrate is the most preferred fatty acid among C2-C6 fatty acids for Sa protein. Like Sa gene transcript, MACS1 mRNA was detected mainly in the liver and kidney. Subcellular fractionation revealed that both MACS1 and Sa protein are localized in the mitochondrial matrix. (14)C-Fatty acid incorporation studies indicated that acyl-CoAs produced by MACS1 and Sa protein are utilized mainly for oxidation.


Subject(s)
Coenzyme A Ligases/genetics , Proteins/genetics , Amino Acid Sequence , Animals , COS Cells , Carbon Radioisotopes , Coenzyme A Ligases/chemistry , Coenzyme A Ligases/isolation & purification , Coenzyme A Ligases/metabolism , DNA, Complementary , Fatty Acids/metabolism , Gene Expression Regulation, Enzymologic , Humans , Mice , Mitochondrial Proteins , Molecular Sequence Data , Proteins/chemistry , Proteins/isolation & purification , Proteins/metabolism , Sequence Homology, Amino Acid , Transfection
5.
Biochem Biophys Res Commun ; 284(4): 993-7, 2001 Jun 22.
Article in English | MEDLINE | ID: mdl-11409893

ABSTRACT

Arachidonate released by various stimuli is rapidly reesterified into membrane phospholipids initiated by acyl-CoA synthetase (ACS) and subsequent acyl-transfer reactions. ACS4 is an arachidonate-preferring enzyme abundant in steroidogenic tissues and postulated to modulate eicosanoid production. Female mice heterozygous for ACS4 deficiency become pregnant less frequently and produce small litters with extremely low transmission of the disrupted alleles. Striking morphological changes, including extremely enlarged uteri and lumina filled with numerous proliferative cysts of various sizes, were detected in ACS4+/- females. Furthermore, marked accumulation of prostaglandins was seen in the uterus of the heterozygous females. These results indicate that ACS4 modulates female fertility and uterine prostaglandin production.


Subject(s)
Coenzyme A Ligases/deficiency , Cysts/genetics , Infertility, Female/genetics , Prostaglandins/metabolism , Uterine Diseases/genetics , Uterus/metabolism , 6-Ketoprostaglandin F1 alpha/metabolism , Animals , Chimera , Coenzyme A Ligases/genetics , Crosses, Genetic , Cysts/enzymology , Cysts/physiopathology , Dinoprost/metabolism , Dinoprostone/metabolism , Female , Genotype , Heterozygote , Infertility, Female/enzymology , Litter Size , Male , Mice , Mice, Knockout , Mice, Transgenic , Restriction Mapping , Sex Ratio , Uterine Diseases/enzymology , Uterine Diseases/physiopathology
6.
J Biol Chem ; 276(14): 11420-6, 2001 Apr 06.
Article in English | MEDLINE | ID: mdl-11150295

ABSTRACT

Using peptide sequences derived from bovine cardiac acetyl-CoA synthetase (AceCS), we isolated and characterized cDNAs for a bovine and murine cardiac enzyme designated AceCS2. We also isolated a murine cDNA encoding a hepatic type enzyme, designated AceCS1, identical to one reported recently (Luong, A., Hannah, V. C., Brown, M. S., and Goldstein, J. L. (2000) J. Biol. Chem. 275, 26458-26466). Murine AceCS1 and AceCS2 were purified to homogeneity and characterized. Among C2-C5 short and medium chain fatty acids, both enzymes preferentially utilize acetate with similar affinity. The AceCS2 transcripts are expressed in a wide range of tissues, with the highest levels in heart, and are apparently absent from the liver. The levels of AceCS2 mRNA in skeletal muscle were increased markedly under ketogenic conditions. Subcellular fractionation revealed that AceCS2 is a mitochondrial matrix enzyme. [(14)C]Acetate incorporation indicated that acetyl-CoAs produced by AceCS2 are utilized mainly for oxidation.


Subject(s)
Acetate-CoA Ligase/analysis , Acetate-CoA Ligase/genetics , Mitochondria/enzymology , Acetate-CoA Ligase/metabolism , Acetates/metabolism , Amino Acid Sequence , Animals , COS Cells , Cattle , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Mice , Molecular Sequence Data , Oxidation-Reduction , Sequence Alignment
7.
Oncogene ; 19(51): 5919-25, 2000 Nov 30.
Article in English | MEDLINE | ID: mdl-11127823

ABSTRACT

Acyl-CoA synthetase (ACS) ligates fatty acid and CoA to produce acyl-CoA, an essential molecule in fatty acid metabolism and cell proliferation. ACS5 is a recently characterized ACS isozyme highly expressed in proliferating 3T3-L1 cells. Molecular characterization of the human ACS5 gene revealed that the gene is located on chromosome 10q25.1-q25.2, spans approximately 46 kb, comprises 21 exons and 22 introns, and encodes a 683 amino acid protein. Two major ACS5 transcripts of 2.5- and 3.7-kb are distributed in a wide range of tissues with the highest expression in uterus and spleen. Markedly increased levels of ACS5 transcripts were detected in a glioma line, A172 cells, and primary gliomas of grade IV malignancy, while ACS5 expression was found to be low in normal brain. Immunohistochemical analysis also revealed strong immunostaining with an anti-ACS5 antibody in glioblastomas. U87MG glioma cells infected with an adenovirus encoding ACS5 displayed induced cell growth on exposure to palmitate. Consistent with the induction of cell growth, the virus infected cells displayed induced uptake of palmitate. These results demonstrate a novel fatty acid-induced glioma cell growth mediated by ACS5.


Subject(s)
Arachidonic Acid/pharmacology , Chromosomes, Human, Pair 10/genetics , Coenzyme A Ligases/genetics , Glioma/enzymology , Palmitic Acid/pharmacology , Adenoviridae/genetics , Adolescent , Adult , Aged , Amino Acid Sequence , Arachidonic Acid/pharmacokinetics , Biological Transport , Carbon Radioisotopes , Cell Division/drug effects , Cell Division/physiology , Coenzyme A Ligases/metabolism , DNA, Complementary/administration & dosage , DNA, Complementary/genetics , Female , Glioma/genetics , Glioma/pathology , Humans , Male , Middle Aged , Molecular Sequence Data , Palmitic Acid/pharmacokinetics , Transfection
8.
Biochem Biophys Res Commun ; 274(3): 741-5, 2000 Aug 11.
Article in English | MEDLINE | ID: mdl-10924347

ABSTRACT

Acyl-CoA synthetase 4 (ACS4) is an arachidonate-preferring enzyme abundant in steroidogenic tissues. We demonstrate that ACS4 expression in steroidogenic tissues in vivo is induced by adrenocorticotropic hormone (ACTH) and suppressed by glucocorticoid. ACTH also induced ACS4 protein but not its mRNA in Y1 adrenocortical tumor cells, whereas both ACS4 mRNA and protein were increased by dibutyryl cAMP (db-cAMP) and forskolin. Furthermore, the levels of ACS4 mRNA and protein in Y1 cells were induced by arachidonate. These data suggest that ACS4 expression in steroidogenic cells is regulated in coordination with induced steroidogenesis and arachidonate released by cholesterol ester hydrolase.


Subject(s)
Adrenocorticotropic Hormone/pharmacology , Arachidonic Acid/pharmacology , Coenzyme A Ligases/biosynthesis , Repressor Proteins , Saccharomyces cerevisiae Proteins , Adrenal Cortex Neoplasms/enzymology , Adrenal Cortex Neoplasms/genetics , Animals , Coenzyme A Ligases/genetics , Female , Gene Expression Regulation, Enzymologic/drug effects , Mice , Mice, Inbred C57BL , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tumor Cells, Cultured
9.
J Biol Chem ; 275(40): 31269-73, 2000 Oct 06.
Article in English | MEDLINE | ID: mdl-10903326

ABSTRACT

Lipoprotein glomerulopathy (LPG) is a unique renal disease characterized by thrombus-like substances in markedly dilated glomerular capillaries, dysbetalipoproteinemia, and elevated plasma concentrations of apoE. Recent studies identified several apoE mutations in patients with LPG, including apoE2(R145P) Sendai (apoE-Sendai). Virus-mediated transduction of apoE-Sendai in apoE-deficient hypercholesterolemic mice resulted in insufficient correction of hypercholesterolemia and a marked and temporal induction of plasma triglyceride levels. In vitro binding studies showed that apoE-Sendai has a reduced affinity for the low density lipoprotein receptor, suggesting that dysbetalipoproteinemia in LPG is caused by the apoE mutation. Furthermore, histological examination revealed marked intraglomerular depositions of apoE-containing lipoproteins in mice injected with apoE-Sendai virus. These LPG-like depositions were detected 6 days after virus injection and were sustained for at least 60 days. Our results demonstrated that apoE-Sendai is an etiological cause of LPG.


Subject(s)
Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Kidney Diseases/etiology , Kidney Diseases/genetics , Respirovirus/genetics , Transduction, Genetic , Adenoviridae/genetics , Animals , Apolipoproteins E/ultrastructure , Chromatography, High Pressure Liquid , Gene Transfer Techniques , Hypercholesterolemia/genetics , Hypercholesterolemia/metabolism , Kidney Diseases/metabolism , Kinetics , Male , Mice , Mice, Mutant Strains , Protein Binding , Receptors, LDL/metabolism , Time Factors , Triglycerides/blood
10.
J Biochem ; 124(6): 1072-6, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9832610

ABSTRACT

The isolation and characterization of rabbit and human cDNAs revealed a new low density lipoprotein receptor (LDLR)-related protein (LRP) designated as LRP5. Human LRP5 cDNA encodes a 1, 616-amino acid type I membrane-like protein with three ligand binding repeats in its extracellular region. LDLR-deficient cells transduced by recombinant adenovirus containing human LRP5 exhibited increased binding of apolipoprotein E (apoE)-enriched beta-migrating very low density lipoprotein. Northern blotting and in situ hybridization revealed a high level of LRP5 expression in hepatocytes and the adrenal gland cortex. In LDLR-deficient Watanabe heritable hyperlipidemic rabbits, LRP5 mRNA was increased in the liver and accumulated in cholesterol-laden foam cells of atherosclerotic lesions.


Subject(s)
Adrenal Cortex/metabolism , Apolipoproteins E/metabolism , Liver/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Adrenal Cortex/chemistry , Amino Acid Sequence , Animals , Apolipoproteins E/chemistry , Arteriosclerosis/metabolism , Binding Sites , Blotting, Northern , Cholesterol/metabolism , Cloning, Molecular , Cytoplasm/metabolism , Humans , Hyperlipidemias/metabolism , In Situ Hybridization/methods , LDL-Receptor Related Proteins , Liver/chemistry , Low Density Lipoprotein Receptor-Related Protein-5 , Male , Molecular Sequence Data , Rabbits , Transcription, Genetic
11.
J Biochem ; 124(4): 747-55, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9756619

ABSTRACT

The very low density lipoprotein receptor (VLDLR) gene contains an exon encoding a region of clustered serine and threonine residues immediately outside the membrane-spanning sequence, and this region has been proposed to be the site of clustered O-linked carbohydrate chains. Two forms of VLDLR transcripts, with and without the O-linked sugar region, are generated through alternative splicing. Reverse transcription polymerase chain reaction with RNAs from various rabbit tissues revealed that the VLDLR transcript with the O-linked sugar region (type-1 VLDLR) is the major transcript in heart and muscle, while the VLDLR transcript without the O-linked sugar region (type-2 VLDLR) predominates in non-muscle tissues, including cerebrum, cerebellum, kidney, spleen, adrenal gland, testis, ovary, and uterus. Hamster fibroblasts expressing type-2 VLDLR bound with relatively low affinity to beta-migrating very low density lipoprotein compared with type-1 VLDLR-transfected cells. In contrast, the internalization, dissociation, and degradation of the ligand were not significantly impaired in either type of VLDLR-transfected cell. The receptor proteins in type-2 VLDLR-transfected cells underwent rapid degradation and accumulated in the culture medium, while those in type-1 VLDLR-transfected cells were stable and resistant to proteolytic cleavage. Analysis of the O-linked sugars of both types of transfected cells suggested that the O-linked sugar region is the major site for O-glycosylation.


Subject(s)
Genetic Variation , Receptors, LDL/chemistry , Receptors, LDL/genetics , Transcription, Genetic , Alternative Splicing , Amino Acid Sequence , Animals , Cell Line , Cricetinae , Female , Glycosylation , Humans , Male , Molecular Sequence Data , Muscle, Skeletal/metabolism , Myocardium/metabolism , Organ Specificity , Protein Processing, Post-Translational , Rabbits , Receptors, LDL/biosynthesis , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Serine , Threonine , Transfection
12.
J Biochem ; 124(4): 784-9, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9756624

ABSTRACT

We report herein the identification of a novel member of the low-density lipoprotein receptor (LDLR) family termed LDLR-related protein 4 (LRP4). Murine LRP4 cDNA encodes a 1113-amino-acid type II membrane-like protein with eight ligand-binding repeats in two clusters. Southern blot analysis of genomic DNA from several different organisms suggests the presence of LRP4 homologues in chicken lacking the gene encoding apolipoprotein E, which is recognized by the ligand-binding repeats of LDLR. LRP4 transcripts were detected almost exclusively in heart in mouse and humans. Despite the presence of the ligand-binding repeats, COS cells transfected with LRP4 did not show surface-binding of beta-migrating very-low-density lipoprotein, suggesting that LRP4 plays a role in a pathway other than lipoprotein metabolism.


Subject(s)
Myocardium/metabolism , Receptors, LDL/chemistry , Receptors, LDL/genetics , Receptors, LDL/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , COS Cells , Chickens , Cloning, Molecular , Consensus Sequence , DNA, Complementary/metabolism , Humans , Kinetics , LDL-Receptor Related Proteins , Mice , Molecular Sequence Data , Receptors, LDL/biosynthesis , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Repetitive Sequences, Amino Acid , Sequence Alignment , Sequence Homology, Amino Acid , Transfection
13.
J Biochem ; 124(3): 679-85, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9722683

ABSTRACT

We report here the identification, characterization, and expression of a novel rat acyl-CoA synthetase (ACS) designated as ACS5. ACS5 consists of 683 amino acids and is approximately 60% identical to the previously characterized ACS1 and ACS2. ACS5 was overproduced in Escherichia coli cells and then purified to near homogeneity. The purified enzyme utilized a wide range of saturated fatty acids similar to those utilized by ACS1 and ACS2, but differed in its preference for C16-C18 unsaturated fatty acids. Northern blot analysis revealed that ACS5 mRNA is present most abundantly in the small intestine, and to a much lesser extent in the lung, liver, adrenal gland, adipose tissue, and kidney. In situ hybridization of rat ileum revealed abundant accumulation of ACS5 transcripts in foveolar epithelial cells. The hepatic level of ACS5 mRNA was significantly increased by refeeding a fat-free high sucrose diet and reduced by fasting or refeeding a high cholesterol diet, whereas that in the small intestine was not significantly altered by various dietary conditions. In contrast to the absence of ACS1 mRNA in undifferentiated 3T3-L1 preadipocytes, ACS5 mRNA was present in proliferating 3T3-L1 preadipocytes and its level remained unaltered during differentiation, suggesting that ACS5 may provide the acyl-CoA utilized for the synthesis of cellular lipids in proliferating preadipocytes.


Subject(s)
Adipocytes/enzymology , Coenzyme A Ligases/metabolism , Intestines/enzymology , Adipocytes/cytology , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Cell Division , Coenzyme A Ligases/genetics , DNA, Complementary , Dietary Fats/administration & dosage , Epithelial Cells/enzymology , Fatty Acids/metabolism , Gene Expression Regulation , In Situ Hybridization , Intestines/cytology , Mitochondrial Proteins , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats
14.
J Biochem ; 124(2): 451-6, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9685741

ABSTRACT

The apolipoprotein E receptor 2 (apoER2) gene consists of a mosaic of exons, which may have been assembled by "exon shuffling." Analysis of apoER2 transcripts in several species reveals a lost repeat in the ligand-binding domain of primate apoER2. A pseudo-exon found in the primate apoER2 genes corresponds to the lost repeat but contains a crucial deletion that leads to a translational frameshift. The pseudo-exon sequence in primary transcripts of the human apoER2 gene is shown to be abolished by exon skipping due to two nucleotide substitutions at the 5'-splice donor adjacent to the pseudo-exon. These data suggest the occurrence of exon loss in the evolution of the primate apoER2 gene.


Subject(s)
Evolution, Molecular , Exons/genetics , Gene Deletion , Receptors, Lipoprotein/genetics , Amino Acid Sequence , Animals , Base Sequence , Callithrix , DNA , Genetic Variation , Humans , LDL-Receptor Related Proteins , Ligands , Mice , Molecular Sequence Data , Repetitive Sequences, Nucleic Acid , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
15.
Genomics ; 51(1): 132-5, 1998 Jul 01.
Article in English | MEDLINE | ID: mdl-9693042

ABSTRACT

We report herein the cDNA cloning of a novel member of the low-density lipoprotein receptor (LDLR) family termed LDLR-related protein 3 (LRP3). Human and rat LRP3 cDNAs encode a 770-amino-acid type 1 membrane protein with the following regions: a putative signal sequence, two isoleucine/leucine/valine-rich regions with an RGD sequence, two ligand-binding repeat regions, a putative transmembrane region, and a proline-rich cytoplasmic region with a tyrosine-based internalization signal. Fluorescence in situ hybridization of human chromosomes revealed that the human LRP3 gene mapped to chromosome bands 19q12-q13.2. LRP3 transcript was detected in a wide range of human tissues, with the highest expression in skeletal muscle and ovary. Despite the presence of the ligand-binding repeats, CHO cells transfected with LRP3 did not bind beta-migrating very-low-density lipoprotein or receptor-associated protein.


Subject(s)
Chromosomes, Human, Pair 19/genetics , Receptors, Immunologic/genetics , Receptors, LDL/genetics , Amino Acid Sequence , Animals , Chromosome Mapping , Cloning, Molecular , DNA, Complementary/genetics , Humans , Low Density Lipoprotein Receptor-Related Protein-1 , Molecular Sequence Data , Rats , Sequence Homology, Amino Acid
16.
Am J Physiol ; 273(1 Pt 1): E37-45, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9252477

ABSTRACT

The effects of sodium cholate on high-fat diet-induced hyperglycemia and obesity were investigated. Insulin resistance was estimated by measuring 2-deoxyglucose uptake in epitrochlearis muscles incubated in vitro. Addition of 0.5% cholate to high-safflower oil diet completely prevented high fat-induced hyperglycemia and obesity in C57BL/6J mice with a slight decrease of energy intake but with no inhibition of fat absorption. Furthermore, the addition of cholate decreased blood insulin levels and prevented high-fat diet-induced decrease of glucose uptake in epitrochlearis. However, there was no change in the unsaturation index of fatty acids in skeletal muscles and in GLUT-4 levels by cholate. In liver, cholate addition resulted in cholesterol accumulation and completely prevented high-fat diet-induced triglyceride accumulation. The changes of triglyceride level in the liver were paralleled to the changes of acyl-CoA synthetase (ACS) mRNA. ACS catalyzes the formation of acyl-CoA from fatty acid, and acyl-CoA is utilized for triglyceride formation in liver. ACS has a sterol-responsive element 1 in its promoter region. These data indicate that the favorable effects of cholate could be partly the result of downregulation of ACS mRNA.


Subject(s)
Cholic Acids/pharmacology , Coenzyme A Ligases/biosynthesis , Dietary Fats , Hyperglycemia/prevention & control , Muscle Proteins , Obesity/prevention & control , RNA, Messenger/biosynthesis , Repressor Proteins , Saccharomyces cerevisiae Proteins , Transcription, Genetic/drug effects , Analysis of Variance , Animals , Cholesterol/metabolism , Cholic Acid , Dietary Fats/analysis , Energy Intake , Fatty Acids/analysis , Fatty Acids/chemistry , Fatty Acids, Nonesterified/metabolism , Feces/chemistry , Female , Gene Expression Regulation, Enzymologic/drug effects , Glucose Transporter Type 4 , Hyperglycemia/etiology , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Monosaccharide Transport Proteins/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Obesity/enzymology , Obesity/etiology , Phospholipids/chemistry , Triglycerides/metabolism
17.
J Biochem ; 122(1): 212-6, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9276691

ABSTRACT

ACS3 is a recently identified acyl-CoA synthetase (ACS) isozyme that preferentially utilizes laurate, myristate, arachidonate, and eicosapentaenoate among saturated and unsaturated long chain fatty acids. The ACS3 purified from COS cells transfected with the ACS3 cDNA was separated by SDS-PAGE into two major forms of 79 and 80 kDa. We report here that alternative translation initiation from ACS3 mRNA gives rise to these two isoforms of ACS3. In vitro mutagenesis of the ACS3 cDNA revealed that the translation of the 80-kDa and 79-kDa isoforms started from the first and second in-frame AUGs, respectively. The two isoforms of ACS3 expressed in COS cells exhibited similar levels of ACS activities toward palmitate and myristate. Immunocytochemistry of intact COS cells transfected with various ACS3 expression vectors suggested that the two forms are localized in the extranuclear compartment, where they exhibit a reticular pattern. In rat cerebrum, the 80-kDa isoform of ACS3 was detected mainly in the microsomal fraction. Only a trace amount of the 79-kDa isoform was detected in rat cerebrum, whereas both forms were detected in rat glioma cell line KEG1 cells.


Subject(s)
Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Protein Biosynthesis , Repressor Proteins , Saccharomyces cerevisiae Proteins , Animals , Brain/metabolism , COS Cells/metabolism , DNA, Complementary , Fatty Acids/metabolism , Immunohistochemistry , Isoenzymes/genetics , Methionine , Mutation , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Nucleic Acid , Subcellular Fractions , Substrate Specificity , Transfection
19.
Proc Natl Acad Sci U S A ; 94(7): 2880-4, 1997 Apr 01.
Article in English | MEDLINE | ID: mdl-9096315

ABSTRACT

We report herein the cDNA cloning of a novel rat acyl-CoA synthetase (ACS) that preferentially uses arachidonate and eicosapentaenoate. This newly identified ACS (designated ACS4) contains 670 amino acids and is 68% identical to rat ACS3, a previously characterized ACS that is highly expressed in brain. ACS4 was overproduced in Escherichia coli and the resulting enzyme was purified to homogeneity. The purified enzyme utilizes arachidonate and eicosapentaenoate most preferentially among C8-C22 saturated fatty acids and C14-C22 unsaturated fatty acids. Kinetic analyses revealed that the enzyme has a high affinity for arachidonate and eicosapentaenoate and low affinity for palmitate. ACS4 transcripts are detectable in a wide range of tissues, with the highest level in adrenal gland. Immunoreactivity to ACS4 was detected in the zona fasciculata and reticularis of adrenal gland, in the corpus luteum and stromal luteinized cells in ovary, and in the Leydig cells of testis.


Subject(s)
Adrenal Glands/enzymology , Arachidonic Acid/metabolism , Coenzyme A Ligases/metabolism , Ovary/enzymology , Repressor Proteins , Saccharomyces cerevisiae Proteins , Testis/enzymology , Amino Acid Sequence , Animals , Blotting, Northern , Coenzyme A Ligases/chemistry , DNA, Complementary , Female , Male , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
20.
J Biol Chem ; 272(13): 8498-504, 1997 Mar 28.
Article in English | MEDLINE | ID: mdl-9079678

ABSTRACT

Apolipoprotein E receptor 2 is a recently identified receptor that resembles low and very low density lipoprotein receptors. Isolation and characterization of genomic clones encoding human apolipoprotein E receptor 2 revealed that the gene spans approximately 60 kilobases and contains 19 exons. The positions of the exon/intron boundaries of the gene are almost identical to those of low and very low density lipoprotein receptors. Fluorescent in situ hybridization of human chromosomes revealed that the gene is located on chromosome 1p34. Isolation of a cDNA encoding a variant receptor and reverse transcription-polymerase chain reaction indicate the presence of multiple variants with different numbers of cysteine-rich repeats in the binding domain of the receptor. We also found a variant receptor lacking a 59-amino acid insertion in the cytoplasmic domain. The transcription start site was mapped to the position 236 base pairs upstream of the AUG translation initiator codon by primer extension analysis. Sequence inspection of the 5'-flanking region revealed potential DNA elements: AP-2, GC factor, PEA3, and Sp1. The minimal promoter region and a region required for nerve growth factor inducibility in PC12 cells were also determined.


Subject(s)
Alternative Splicing , Apolipoproteins/genetics , Exons , Introns , Receptors, Lipoprotein/genetics , Animals , Base Sequence , Chromosome Mapping , Cricetinae , Cricetulus , Humans , In Situ Hybridization, Fluorescence , LDL-Receptor Related Proteins , Low Density Lipoprotein Receptor-Related Protein-1 , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...