Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
J Mol Endocrinol ; 34(1): 237-45, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15691891

ABSTRACT

The enzyme 25-hydroxyvitamin D 1alpha-hydroxylase, or CYP27B1, is the key enzyme in the two-step activation process of vitamin D to 1,25-dihydroxyvitamin D (1,25D). While a number of regulators of the renal CYP27B1 enzyme activity have been recognized for some years, their underlying molecular mechanisms remain largely unknown, and the DNA regions involved in the in vivo regulation of gene expression by these factors have not been delineated. We have generated a transgenic mouse line that expresses 1501 bp of 5' flanking region together with 44 bp of 5' untranslated region of the human CYP27B1 gene fused to the firefly luciferase reporter gene. Animals expressing the luciferase gene demonstrated that both luciferase protein and mRNA for CYP27B1 were localized to proximal convoluted tubule cells of the kidney. In 2-week-old animals, the expression of the transgene and the endogenous CYP27B1 mRNA levels in the kidney were highest and fell with increasing age. Both reporter gene expression and CYP27B1 mRNA levels were downregulated in response to increasing amounts of dietary calcium in a dose-dependent manner. Vitamin D deficiency resulted in an increase in both the reporter gene and CYP27B1 expression. Interestingly, the increase in CYP27B1 mRNA levels was substantially higher than the increase in reporter gene expression, suggesting either that there is a post-transcriptional mechanism that increases the amount of CYP27B1 mRNA or that other regulatory elements are required to maximize the effect of vitamin D deficiency. These findings demonstrate that the 1501 bp 5' flanking region of the CYP27B1 gene directs expression to the proximal convoluted tubules of the kidney and is responsible for increasing transcriptional activity when dietary calcium and vitamin D levels are depleted. It also responds in the kidney to the physiological regulators of development and ageing.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , 5' Flanking Region , Gene Expression Regulation/physiology , Promoter Regions, Genetic , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/biosynthesis , Age Factors , Animals , Calcium/metabolism , Genes, Reporter , Immunohistochemistry , Kidney/metabolism , Mice , Mice, Transgenic , Vitamin D Deficiency/metabolism
2.
J Steroid Biochem Mol Biol ; 89-90(1-5): 159-62, 2004 May.
Article in English | MEDLINE | ID: mdl-15225765

ABSTRACT

25-hydroxyvitamin D(3)- or 1alpha,25-dihydroxyvitamin D(3)-24R-hydroxylase (cytochromeP450C24 or CYP24) has a dual role of removing 25-OH-D(3) from circulation and excess 1,25(OH)(2)D(3) from kidney. As a result, CYP24 is an important multifunctional regulatory enzyme that maintains essential tissue-levels of Vitamin D hormone. As a part of our continuing interest in structure-function studies characterizing various binding proteins in the Vitamin D endocrine system, we targeted recombinant rat CYP24 with a radiolabeled 25-OH-D(3) affinity analog, and showed that the 25-OH-D(3)-binding site was specifically labeled by this analog. An affinity labeled sample of CYP24 was subjected to MS/MS analysis, which identified Ser57 as the only amino acid residue in the entire length of the protein that was covalently modified by this analog. Site-directed mutagenesis was conducted to validate the role of Ser57 towards substrate-binding. S57A mutant displayed significantly lower binding capacity for 25-OH-D(3) and 1,25(OH)(2)D(3). On the other hand, S57D mutant strongly enhanced binding for the substrates and conversion of 1,25(OH)(2)D(3) to calcitroic acid. The affinity probe was anchored via the 3-hydroxyl group of 25-OH-D(3). Therefore, these results suggested that the 3-hydroxyl group (of 25-OH-D(3) and 1,25(OH)(2)D(3)) in the S57D mutant could be stabilized by hydrogen bonding or a salt bridge leading to enhanced substrate affinity and metabolism.


Subject(s)
Affinity Labels , Cytochrome P-450 Enzyme System/chemistry , Serine/chemistry , Steroid Hydroxylases/chemistry , Animals , Binding Sites , Cytochrome P-450 Enzyme System/metabolism , Mass Spectrometry , Rats , Serine/metabolism , Steroid Hydroxylases/metabolism , Vitamin D3 24-Hydroxylase
3.
J Steroid Biochem Mol Biol ; 89-90(1-5): 409-12, 2004 May.
Article in English | MEDLINE | ID: mdl-15225811

ABSTRACT

The aim of this study was to investigate effects of 1,25(OH)(2)D(3) (calcitriol), 25OHD(3), and EB1089 on cell growth and on Vitamin D receptor (VDR) mRNA and 1alpha-hydroxylase (1alpha-OHase) mRNA expression in normal canine prostatic primary cultures. Canine prostatic epithelial cells were isolated, cultured, and treated with vehicle (ethanol), calcitriol, 25OHD(3), and EB1089 at 10(-9) and 10(-7)M. The VDR was present in epithelial and stromal cells of the canine prostate gland. 1,25(OH)(2)D(3), 25OHD(3), and EB1089 inhibited epithelial cell growth at 10(-7)M compared to vehicle-treated controls [calcitriol (P < 0.01), EB1089 (P < 0.01), and 25OHD(3) (P < 0.05)]. Epithelial cells treated with calcitriol and EB1089 at 10(-7)M had slightly increased VDR mRNA expression (0.2-0.3-fold) at 6 and 12h compared to controls. There was no difference in 1alpha-OHase mRNA expression in epithelial cells treated with these three compounds. 1,25(OH)(2)D(3) and its analogs may be effective antiproliferative agents of epithelial cells in certain types of prostate cancer.


Subject(s)
Calcifediol/pharmacology , Calcitriol/pharmacology , Cell Division/drug effects , Prostate/drug effects , RNA, Messenger/genetics , Receptors, Calcitriol/genetics , Steroid Hydroxylases/genetics , Animals , Cells, Cultured , Dogs , Immunohistochemistry , Male , Prostate/cytology , Prostate/enzymology , Prostate/metabolism , RNA, Messenger/metabolism
4.
J Mol Endocrinol ; 32(1): 87-98, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14765994

ABSTRACT

Regulation of the gene for renal 25-hydroxyvitamin D-24-hydroxylase (CYP24) is important for controlling the level of circulating 1,25-dihydroxyvitamin D3 (1,25(OH)2D3). We report here for the first time that the peptide hormone calcitonin significantly stimulates expression of a rat CYP24 promoter-luciferase construct in both transiently and stably transfected kidney HEK-293 cells. A GC box at -114/-101 and a CCAAT box at -62/-51 have been identified that underlie both basal expression of the CYP24 promoter and the calcitonin inductive response. Data from overexpression studies suggested that Sp1 and NF-Y are the proteins that function through the GC and CCAAT boxes respectively. ERK1/2 signaling pathways were not involved in the calcitonin-mediated response, since stimulation of the promoter was unaffected by the pharmacological ERK1/2 inhibitor PD98059 and by a dominant negative mutant of ERK1/2 (ERK1K71R). In contrast, calcitonin induction but not basal expression was dependent on protein kinase A and protein kinase C (PKC) activities with the inhibitors H89 and calphostin C lowering induction by 50-60%. The atypical PKC, PKCzeta contributes to calcitonin induction, but not to basal expression of the CYP24 promoter, since overexpression of a dominant negative clone PKCzetaK281 M lowered induction by 50%. Cotransfection of a dominant negative form of Ras resulted in calcitonin-mediated induction being reduced also by about 50%. A Ras-PKCzeta signaling pathway for calcitonin action is proposed, which acts through the GC box. The findings have been extrapolated to the in vivo situation where we suggest that induction of renal CYP24 by calcitonin could be important under hypercalcemic conditions thus contributing to the lowering of circulating 1,25(OH)2D3 levels.


Subject(s)
Calcitonin/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytochrome P-450 Enzyme System/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Receptors, Calcitonin/metabolism , Steroid Hydroxylases/metabolism , Animals , CCAAT-Binding Factor/metabolism , Cells, Cultured , Cloning, Molecular , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Flavonoids/pharmacology , Genes, Reporter/genetics , Humans , Immunoglobulins/metabolism , Isoquinolines/pharmacology , Mutation , Naphthalenes/pharmacology , Promoter Regions, Genetic/genetics , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Sulfonamides/pharmacology , Vitamin D3 24-Hydroxylase
5.
J Nutr Biochem ; 14(7): 378-85, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12915218

ABSTRACT

The molecular basis for pseudo vitamin D deficiency rickets (PDDR) in the Hannover pig model was determined in the current study. Consistent with the inability of Hannover PDDR pigs to maintain ambient levels of 1,25-dihydroxyvitamin D (i.e., 1,25D), the bioactivation enzyme cytochrome P450C1 (or CYP27B1) was determined to contain coding-region deletions that rendered the enzyme ineffective due to frame-shift mutations and expression of a premature termination codon. Expression levels of P450C1mRNA were up-regulated in response to the low-1,25D high-parathyroid hormone state of the PDDR animals. In a complementary manner, cytochrome P450C24 mRNA was not detectable in PDDR pigs. Two different deletions were detected within the Hannover pig strain in which the P450C1 coding region contained either 173 bp or 329 bp deletions that resulted in the expression of non-sense products beginning within the I-helix region and extending through the truncated C-terminal domains. The boundaries for the deletion segments aligned with derived mRNA processing sites. This observation was consistent with an mRNA processing error as the causative factor for the coding-region deletions. Based upon the expression of a non-functional P450C1 enzyme, the Hannover pig model for PDDR was determined to be identical to the human disease in which enzyme-inhibitory mutations are the molecular basis for the calcium disorder.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , Hypophosphatemia, Familial/genetics , Vitamin D Deficiency , Animals , Base Sequence , Cloning, Molecular , DNA/chemistry , Gene Deletion , Gene Expression , Humans , Kidney/enzymology , Mice , Molecular Sequence Data , Mutation , RNA, Messenger/analysis , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Swine
6.
J Cell Biochem ; 88(2): 356-62, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12520537

ABSTRACT

Although vitamin D(3) is a natural product of a sunlight-mediated process in the skin, the secosteroid's biological function is dependent upon specific cytochrome P450 enzymes that mediate the parent vitamin's bioactivation and inactivation. Cytochrome P450C1 (CYP27B1) is the regulatory rate-limiting enzyme that directs the bioactivation process through introduction of a C-1alpha hydroxyl group. The resultant 1,25-dihydroxyvitamin D(3) (1,25D) is the biologically active secosteroid hormone that directs the multitude of vitamin D-dependent actions involved with calcium homeostasis, cellular differentiation and growth, and the immune response. The circulating and cellular level of 1,25D is regulated through a coordinated process involving the hormone's synthesis and degradation. Central to the degradation and turnover of 1,25D is the regulatory multi-catalytic cytochrome P450C24 (CYP24) enzyme that directs the introduction of C-24R groups onto targeted 25-hydroxy substrates. Discussed in this article is the action of the rat CYP24 to catalyze the side-chain oxidation and cleavage of 25-hydroxylated vitamin D metabolites. Expression and characterization of purified recombinant rat CYP24 is discussed in light of mutations directed at the enzyme's active site.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase , Cytochrome P-450 Enzyme System , Models, Molecular , Steroid Hydroxylases , Vitamin D/metabolism , Animals , Humans , Structure-Activity Relationship , Vitamin D3 24-Hydroxylase
7.
Steroids ; 66(3-5): 381-9, 2001.
Article in English | MEDLINE | ID: mdl-11179747

ABSTRACT

Cytochromes P450c1 and P450c24 are regulated hydroxylase enzymes that direct the bioactivation and metabolic degradation of vitamin D. The bioactivation pathway is regulated by cytochrome P450c1 through its synthesis of 1alpha,25(OH)(2)D(3), the hormonally active form of the vitamin. Expression of the P450c1 gene is regulated at the transcription level. Promoter regions within the P450c1 gene have been identified that respond to cAMP and 1alpha,25(OH)(2)D(3) during the respective up- and down-regulation of P450c1 gene expression. The diametric action of 1alpha,25(OH)(2)D(3) to up-regulate P450c24 gene expression is discussed in the context of two vitamin D response elements (VDREs) that are linked functionally to an adjoining Ets-binding site. It is apparent from sequence-derived data that the P450c1 and P450c24 enzymes share only 10-25% sequence identity, yet they display functionally similar domains that are conserved across the family of cytochrome P450 enzymes. Expression of E. coli recombinant P450c1 and P450c24 enzymes, and the substrate-binding parameters for P450c24 are discussed. Finally, the natural point mutations in human P540c1 from patients with pseudovitamin D-deficiency rickets (PDDR) are discussed in the context of the enzyme's structure and function.


Subject(s)
Steroid Hydroxylases/genetics , Vitamin D/metabolism , Amino Acid Sequence , Animals , Calcitriol/biosynthesis , Cytochrome P-450 Enzyme System/metabolism , Gene Expression Regulation , Humans , Molecular Sequence Data , Sequence Alignment , Steroid Hydroxylases/metabolism , Steroid Hydroxylases/physiology , Vitamin D/physiology
8.
J Biol Chem ; 275(1): 47-55, 2000 Jan 07.
Article in English | MEDLINE | ID: mdl-10617584

ABSTRACT

Transcription of the rat CYP24 gene is induced by 1, 25-dihydroxyvitamin D(3) (1,25-(OH)(2)D(3)) through two vitamin D response elements (VDREs). A functional Ras-dependent Ets-binding site (EBS) was located downstream from the proximal VDRE and was critical to 1,25(OH)(2)D(3)-mediated induction. Cotransfection of Ets-1 and Ets-2 stimulated induction, which was lost when the EBS was mutated. Multiple nuclear-protein complexes from COS-1 cells bound to the EBS in which three complexes were immunologically related to Ets-1. Transcriptional synergy was observed between the proximal VDRE and adjacent EBS as was the attendant formation of a ternary complex between vitamin D receptor- retinoid X receptor (VDR. RXR) and Ets-1. In the absence of 1,25-(OH)(2)D(3) or in the presence of an inactive proximal VDRE, the EBS failed to respond to exogenous Ets-1. However, Ets-1 increased basal expression when cotransfected with a mutant VDR. The inductive action of 1, 25-(OH)(2)D(3) was substantially increased by Ras, which was ablated by mutagenesis of the EBS or by expression of a mutated Ets-1 protein (T38A). EBS contribution to hormone induction was prevented by manumycin A, an inhibitor of Ras farnesylation. A fundamental role was established for transcriptional cooperation between Ras-activated Ets proteins and the VDR.RXR complex in mediating 1, 25-(OH)(2)D(3) action on the CYP24 promoter.


Subject(s)
Calcitriol/pharmacology , Cytochrome P-450 Enzyme System/genetics , Proto-Oncogene Proteins/metabolism , Receptors, Calcitriol/metabolism , Response Elements , Steroid Hydroxylases/genetics , Transcription Factors/metabolism , Transcriptional Activation , ras Proteins/metabolism , Animals , Binding Sites , Cytochrome P-450 Enzyme System/biosynthesis , Dimerization , Gene Expression Regulation, Enzymologic , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Protein c-ets-1 , Proto-Oncogene Proteins c-ets , Proto-Oncogene Proteins c-raf/metabolism , Rats , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors , Steroid Hydroxylases/biosynthesis , Transcription Factor AP-1/metabolism , Vitamin D3 24-Hydroxylase
9.
Am J Physiol ; 276(4): E793-805, 1999 04.
Article in English | MEDLINE | ID: mdl-10198318

ABSTRACT

Previous studies using microdissected nephron segments reported that the exclusive site of renal 25-hydroxyvitamin D3-24-hydroxylase (24OHase) activity is the renal proximal convoluted tubule (PCT). We now report the presence of 24OHase mRNA, protein, and activity in cells that are devoid of markers of proximal tubules but express characteristics highly specific for the distal tubule. 24OHase mRNA was undetectable in vehicle-treated mouse distal convoluted tubule (DCT) cells but was markedly induced when DCT cells were treated with 1,25 dihydroxyvitamin D3 [1,25(OH)2D3]. 24OHase protein and activity were also identified in DCT cells by Western blot analysis and HPLC, respectively. 8-Bromo-cAMP (1 mM) or parathyroid hormone [PTH-(1-34); 10 nM] was found to potentiate the effect of 1, 25(OH)2D3 on 24OHase mRNA. The stimulatory effect of cAMP or PTH on 24OHase expression in DCT cells suggests differential regulation of 24OHase expression in the PCT and DCT. In the presence of cAMP and 1, 25(OH)2D3, a four- to sixfold induction in vitamin D receptor (VDR) mRNA was observed. VDR protein, as determined by Western blot analysis, was also enhanced in the presence of cAMP. Transient transfection analysis in DCT cells with rat 24OHase promoter deletion constructs demonstrated that cAMP enhanced 1, 25(OH)2D3-induced 24OHase transcription but this enhancement was not mediated by cAMP response elements (CREs) in the 24OHase promoter. We conclude that 1) although the PCT is the major site of localization of 24OHase, 24OHase mRNA and activity can also be localized in the distal nephron; 2) both PTH and cAMP modulate the induction of 24OHase expression by 1,25(OH)2D3 in DCT cells in a manner different from that reported in the PCT; and 3) in DCT cells, upregulation of VDR levels by cAMP, and not an effect on CREs in the 24OHase promoter, is one mechanism involved in the cAMP-mediated modulation of 24OHase transcription.


Subject(s)
Calcitriol/pharmacology , Cyclic AMP/physiology , Cytochrome P-450 Enzyme System , Gene Expression Regulation, Enzymologic/physiology , Kidney Tubules/enzymology , Nephrons/enzymology , Parathyroid Hormone/physiology , Steroid Hydroxylases/genetics , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Base Sequence , Cells, Cultured , Cyclic AMP/pharmacology , Enzyme Induction , Gene Expression Regulation, Enzymologic/drug effects , Kidney Tubules, Distal/enzymology , Mice , Molecular Sequence Data , RNA, Messenger/genetics , Rats , Recombinant Fusion Proteins/biosynthesis , Regulatory Sequences, Nucleic Acid , Steroid Hydroxylases/biosynthesis , Teriparatide/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic/drug effects , Transfection , Vitamin D3 24-Hydroxylase
10.
Am J Physiol ; 276(2): H699-708, 1999 02.
Article in English | MEDLINE | ID: mdl-9950873

ABSTRACT

We previously demonstrated augmented endothelium-derived nitric oxide (EDNO)-dependent pulmonary arterial dilation and increased arterial endothelial nitric oxide synthase (eNOS) levels in chronic hypoxic (CH) and monocrotaline (nonhypoxic) models of pulmonary arterial hypertension. Therefore, we hypothesized that the long-term elevation of arterial eNOS levels associated with CH is related to pulmonary hypertension or some factor(s) associated with hypertension and not directly to hypoxia. To test this hypothesis, we examined responses to the EDNO-dependent dilator ionomycin in U-46619-constricted, isolated, saline-perfused lungs from control rats, CH (4 wk at 380 mmHg) rats, and rats previously exposed to CH but returned to normoxia for 4 days or 2 wk. Microvascular pressure was assessed by double-occlusion technique, allowing calculation of segmental resistances. In addition, vascular eNOS immunoreactivity was assessed by quantitative immunohistochemistry, and eNOS mRNA abundance was determined by RT-PCR assays. Our findings indicate that 4-day and 2-wk posthypoxic rats exhibit persistent pulmonary hypertension, likely due to maintained arterial remodeling and polycythemia associated with prior exposure to CH. Furthermore, arterial dilation to ionomycin was augmented in lungs from each experimental group compared with controls. Finally, arterial eNOS immunoreactivity and whole lung eNOS mRNA levels remained elevated in posthypoxic animals. These findings suggest that altered vascular mechanical forces or vascular remodeling contributes to enhanced EDNO-dependent arterial dilation and upregulation of arterial eNOS in various models of established pulmonary hypertension.


Subject(s)
Gene Expression Regulation/physiology , Hypoxia/enzymology , Hypoxia/genetics , Lung/enzymology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Chronic Disease , Gases/blood , Hematocrit , Hemodynamics/physiology , Hypertrophy, Right Ventricular/etiology , Hypoxia/complications , Hypoxia/physiopathology , In Vitro Techniques , Ionomycin/pharmacology , Male , Nitric Oxide Synthase Type III , Pulmonary Circulation/drug effects , Pulmonary Circulation/physiology , Rats , Rats, Sprague-Dawley , Vascular Resistance/drug effects , Vascular Resistance/physiology
11.
Alcohol Clin Exp Res ; 23(12): 1868-74, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10630604

ABSTRACT

BACKGROUND: Monitoring CYP2E1 levels in alcoholic individuals holds inherent appeal because such determinations might indicate individuals at increased risk for alcoholic liver disease. We previously demonstrated that lymphocyte CYP2E1 expression reflects in vivo activity of the hepatic enzyme. METHODS: To further validate this approach, the current investigation compared lymphocyte CYP2E1 content and chlorzoxazone pharmacokinetics in 51 alcoholic and nonalcoholic White, Navajo, and Mexican American subjects. After an oral dose of chlorzoxazone, blood samples were collected and lymphocytes isolated. RESULTS: Alcoholics exhibited a 2-fold elevation in lymphocyte CYP2E1 messenger ribonucleic acid (mRNA) and protein compared to nonalcoholics. Chlorzoxazone clearance rates were 1.9-fold higher and area under the concentration curve (AUC) values 1.8-fold lower in alcoholic individuals compared to nonalcoholics. Furthermore, chlorzoxazone clearance rates correlated (r = 0.55, p < 0.01, n = 38) with lymphocyte CYP2E1 mRNA content, and transcript levels further correlated (r = 0.52, p < 0.001, n = 38) with CYP2E1 protein content. To compare phenotype with genotype, restriction fragment length polymorphism analyses on deoxyribonucleic acid samples were performed to identify polymorphisms in the CYP2E1 gene. No subjects were homozygous for rare alleles c2 or C. Nonetheless, 27% of the Navajos and 15% of the Mexican Americans were heterozygous for the c2 allele. Two White subjects appeared heterozygous (c1/c2) when RsaI was used to characterize CYP2E1 genotype but homozygous (c1/c1) at the PstI locus. Fifteen percent of Mexican American subjects, 20% of Navajo subjects, and 6% of White subjects were heterozygous for the C allele. Neither CD nor cl/c2 genotypes were associated with alcoholism. CONCLUSIONS: Human lymphocyte CYP2E1 mRNA levels may be useful predictors of alcohol-mediated alterations in hepatic CYP2E1 activity. Moreover, ethnicity does not appear to play a major role in the levels of expression of lymphocyte CYP2E1.


Subject(s)
Alcoholism/blood , Chlorzoxazone/pharmacokinetics , Cytochrome P-450 CYP2E1/blood , Lymphocytes/enzymology , Muscle Relaxants, Central/pharmacokinetics , RNA, Messenger/blood , Adult , Alcoholism/genetics , Alleles , Cytochrome P-450 CYP2E1/genetics , Female , Humans , Indians, North American/genetics , Male , Mexican Americans/genetics , Middle Aged , RNA, Messenger/genetics , White People/genetics
12.
J Mol Endocrinol ; 20(3): 327-35, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9687155

ABSTRACT

Repression of basal transcription of a 1,25-dihydroxyvitamin D3 (1,25-(OH)2D3) responsive 25-hydroxyvitamin D3-24-hydroxylase (CYP24) promoter construct as observed in kidney cells in the absence of ligand and this repression was dependent on a functional vitamin D response element (VDRE). Basal repression was also seen with a construct where a consensus DR-3-type VDRE was fused to the thymidine kinase promoter. Expression of a dominant negative vitamin D receptor (VDR) isoform that strongly bound to the VDRE motif in the CYP24 promoter ablated basal repression. This VDR isoform lacked sequence in the hinge- and ligand-binding domains implicating one or both of these domains in basal repression. It is well known that thyroid hormone and retinoic acid receptors silence basal transcription of target genes in the absence of ligands and this repressor function can be mediated by the nuclear receptor corepressor N-CoR. Two variants of N-CoR have been described, RIP13a and RIP13delta1. N-CoR and the variants contain two receptor interaction domains, ID-I and ID-II, which are identical except region ID-II in RIP13delta1 has an internal deletion. We have used the mammalian two hybrid system to investigate whether VDR, in the absence of ligand 1,25-(OH)2D3, can interact with these domains. The data showed that unliganded VDR does not interact with either ID-I or ID-II from RIP13a and RIP13delta1, but does interact strongly with a composite domain of ID-I and ID-II from RIP13delta1 (but not from RIP13a) and this strong interaction is abrogated in the presence of ligand. This finding implicates RIP13delta1 in VDR-dependent basal repression of the promoter constructs under investigation. However, over-expression of RIP13delta1 in kidney cell lines did not alter basal expression of the CYP24 promoter construct. It is concluded that either the level of endogenous RIP13delta1 in these kidney cells permits maximal repression or that repression occurs by a mechanism that is independent of RIP13delta1. Alternatively, repression may be dependent on RIP13delta1 but requires an additional cofactor that is limiting in these cells.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Genetic Variation , Kidney/enzymology , Promoter Regions, Genetic , Receptors, Calcitriol/metabolism , Steroid Hydroxylases/genetics , Transcription, Genetic , Animals , Base Sequence , Binding Sites , COS Cells , Calcitriol/pharmacology , Cell Line , Consensus Sequence , Cytochrome P-450 Enzyme System/biosynthesis , DNA Primers , Enzyme Repression , Humans , Recombinant Fusion Proteins/biosynthesis , Restriction Mapping , Steroid Hydroxylases/biosynthesis , Transfection , Vitamin D3 24-Hydroxylase
13.
J Bone Miner Res ; 12(10): 1552-9, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9333115

ABSTRACT

Pseudovitamin D-deficiency rickets (PDDR) is an autosomal recessive disorder that may be due to impaired activity of 25-hydroxyvitamin D-1alpha-hydroxylase, a renal cytochrome P450 enzyme (P450[1alpha]) of the vitamin D pathway. The disease locus for PDDR has been mapped by linkage analysis to 12q13-q14, but the molecular defect underlying the enzyme dysfunction has remained elusive due to the lack of sequence information for the P450(1alpha) gene (hereafter referred to as 1alpha-OHase). We have used a probe derived from the rat 25-hydroxyvitamin D-24-hydroxylase (CYP24; 24-OHase) sequence to identify and clone the 1alpha-OHase cDNA. The full-length 1alpha-OHase clone of 2.4 kb codes for a protein of predicted Mr 55 kDa. Functional activity of the cloned sequence was assessed using transient transfection, and the production of authentic 1alpha,25-dihydroxyvitamin D3 [1alpha,25(OH)2D3] was confirmed using high performance liquid chromatography fractionation and time-of-flight mass spectrometry. The expression of the gene was analyzed in vitamin D-replete animals; treatment with 1alpha,25(OH)2D3 reduced 1alpha-OHase transcript levels by 70%, while administration of parathyroid hormone led to a 2-fold increase in the expression of the gene, thus confirming the hormonal regulation previously described using biochemical methods. The rat cDNA was used to obtain a human genomic clone. Interestingly, the human 1alpha-OHase gene mapped to 12q13.1-q13.3, providing strong evidence that a mutation in the 1alpha-OHase gene is responsible for the PDDR phenotype. The availability of a cloned sequence for 1alpha-OHase generates novel tools for the study of the molecular etiology of PDDR, and will allow the investigation of other disturbances of vitamin D metabolism.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , Cytochrome P-450 Enzyme System/genetics , Rickets/genetics , Steroid Hydroxylases/genetics , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/chemistry , Amino Acid Sequence , Animals , Calcitriol/chemistry , Calcitriol/therapeutic use , Cloning, Molecular , Cytochrome P-450 Enzyme System/chemistry , DNA, Complementary/chemistry , DNA, Complementary/genetics , Gene Expression Regulation, Enzymologic/genetics , Humans , Molecular Sequence Data , Molecular Weight , Parathyroid Hormone/therapeutic use , Rats , Rats, Sprague-Dawley , Restriction Mapping , Rickets/enzymology , Rickets/etiology , Sequence Homology, Amino Acid , Steroid Hydroxylases/chemistry , Transcription, Genetic/genetics , Transfection , Vitamin D Deficiency/enzymology , Vitamin D Deficiency/genetics , Vitamin D3 24-Hydroxylase
14.
Drug Metab Dispos ; 25(12): 1429-35, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9394034

ABSTRACT

Cytochrome P450 (CYP) 2E1 is implicated in a variety of chemically initiated hepatotoxicities, including alcoholic liver disease. These pathological conditions arise from increased production of reactive intermediates caused by elevated enzyme concentrations. Thus, the ability to detect enhanced CYP2E1 levels would aid in identifying individuals at high risk for xenobiotic-promoted liver injury. With this in mind, the present investigation assessed in vivo chlorzoxazone metabolism and compared pharmacokinetic parameters with CYP2E1 expression in blood. Twenty-two subjects were recruited and divided into two groups, control subjects and alcohol abusers, based on responses to two screening questionnaires. Those individuals with higher survey scores, i.e. those who consumed alcohol more frequently, exhibited higher rates of chlorzoxazone metabolism. Indeed, a correlation (r = 0.66, p < 0.01) was obtained when scores were compared with the pharmacokinetic parameter AUC for chlorzoxazone. Lymphocyte microsomes isolated from blood samples obtained from these same individuals were subjected to immunoblot analyses to detect CYP2E1 levels. That lymphocytes contained CYP2E1 was confirmed by reverse transcription-polymerase chain reaction and sequence analysis of the cDNA. Quantification of immunoreactive bands revealed that levels of this P450 were 2.3-fold higher in alcoholics than in control subjects. This increase in lymphocyte CYP2E1 content in alcoholic subjects coincided with a 2.1-fold increase in chlorzoxazone clearance and a 2-fold decrease in the AUC for chlorzoxazone. Importantly, a correlation (r = 0.62, p < 0.01) was observed between CYP2E1 content in lymphocytes and chlorzoxazone clearance rates. Thus, monitoring lymphocyte CYP2E1 expression may provide a substitute for estimating hepatic activity of this P450.


Subject(s)
Chlorzoxazone/metabolism , Cytochrome P-450 CYP2E1/drug effects , Ethanol/pharmacology , Liver/drug effects , Liver/enzymology , Lymphocytes/drug effects , Lymphocytes/enzymology , Adult , Biomarkers/blood , Cytochrome P-450 CYP2E1/blood , Female , Humans , Male , Middle Aged , Random Allocation
15.
J Biol Chem ; 271(47): 29715-21, 1996 Nov 22.
Article in English | MEDLINE | ID: mdl-8939905

ABSTRACT

Transcription of the CYP24 gene is induced by 1,25-(OH)2D3 through a vitamin D receptor-dependent process. The functional activities of three possible vitamin D response elements (VDREs), located on the antisense strand of the rat CYP24 promoter, were investigated by transient expression of native and mutant promoter constructs in COS-1, JTC-12, and ROS 17/2.8 cells. A putative VDRE with a half-site spacing of 6 base pairs at -249/-232 (VDRE-3) did not contribute to 1,25-(OH)2D3 induced expression in the native promoter, although activity has been reported when the element was fused to the heterologous thymidine kinase promoter. Two VDREs with half-site spacings of 3 base pairs at -150/-136 and -258/-244 (VDRE-1 and VDRE-2, respectively), showed transcriptional synergism in COS-1 cells when treated with 1,25-(OH)2D3 (10(-7) to 10(-11) M). The contribution of both VDREs was hormone-concentration dependent from 10(-10) to 10(-12) M, with VDRE-1 demonstrating greatest sensitivity to 1,25-(OH)2D3. Transactivation by VDRE-1 was always greater than VDRE-2, but the converse was observed for the binding of vitamin D receptor-retinoid X receptor complex by each VDRE in gel mobility shift assays. The synergy observed between VDRE-1 and VDRE-2 may have important implications in cellular responses to different circulating levels of 1,25-(OH)2D3.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Promoter Regions, Genetic , Steroid Hydroxylases/genetics , Transcription, Genetic , Vitamin D/metabolism , Animals , Cell Line , Cytochrome P-450 Enzyme System/metabolism , Gene Expression Regulation, Enzymologic , Mutagenesis, Site-Directed , Nuclear Proteins/metabolism , Protein Binding , Rats , Steroid Hydroxylases/metabolism , Vitamin D3 24-Hydroxylase
16.
Proc Soc Exp Biol Med ; 207(1): 57-61, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7938037

ABSTRACT

It is well documented that 1 alpha,25-dihydroxyvitamin D3 (1 alpha,25[OH]2D3), the most active vitamin D metabolite, inhibits epidermal keratinocyte proliferation and promotes differentiation. 1 alpha,25(OH)2D3 can be produced in keratinocytes from 25-hydroxyvitamin D3 by the enzyme 25-hydroxyvitamin D3-1 alpha-hydroxylase (1-OHase). Hydroxylation of 1 alpha,25(OH)2D3 by 25-hydroxyvitamin D3-24-hydroxylase (24-OHase), the first step in the catabolic pathway of 1 alpha,25(OH)2D3 could significantly reduce the intracellular concentration of 1 alpha,25(OH)2D3. Therefore, the expression of 24-OHase could have a critical regulatory role in 1 alpha,25(OH)2D3-dependent gene expression. As a first step to examine this possibility, the steady state level of 24-OHase mRNA in cultured human keratinocytes (CHK) was investigated. 24-OHase mRNA was not detected in control CHK. 1 alpha,25(OH)2D3 caused a dose- and time-dependent increase in 24-OHase mRNA level. The highest accumulation of 24-OHase mRNA was observed in CHK treated with 0.1-1 microM 1 alpha,25(OH)2D3. The level of 24-OHase mRNA reached a plateau 12-24 hr after 1 alpha,25(OH)2D3 treatment. 1 beta,25-dihydroxyvitamin D3, the stereoisomer of 1 alpha,25(OH)2D3, failed to induce 24-OHase mRNA expression significantly. In addition to 24-OHase mRNA, a 1.0-kb mRNA hybridized strongly with both rat and human 24-OHase cDNA probes. The origin of this 1.0-kb message is unknown at present, however, it was regulated by 1 alpha,25(OH)2D3. These results demonstrate that 1 alpha,25(OH)2D3 up-regulates the expression of 24-OHase mRNA, and this may be an important first step in the initiation of catabolism of 1 alpha,25(OH)2D3 in human keratinocytes.


Subject(s)
Calcitriol/pharmacology , Cytochrome P-450 Enzyme System , Keratinocytes/metabolism , Steroid Hydroxylases/genetics , Cells, Cultured , Enzyme Induction/drug effects , Gene Expression/drug effects , Humans , In Vitro Techniques , Male , RNA, Messenger/genetics , Vitamin D3 24-Hydroxylase
17.
Nucleic Acids Res ; 22(12): 2410-6, 1994 Jun 25.
Article in English | MEDLINE | ID: mdl-8036172

ABSTRACT

Mitochondrial cytochrome P450(24) expression in the vitamin D-degradation pathway is induced by 1,25-dihydroxyvitamin D3 [1,25-(OH)2D3]. The molecular basis of this enzyme regulation was investigated by isolating the rat P450(24) gene and examining the 5'-flanking region for possible cis-acting regulatory elements involved in the induction process. Constructs containing different lengths of 5'-flanking region of the gene were linked to a luciferase reporter gene and transiently co-transfected with a human vitamin D receptor (hVDR) expression vector (pRSV-hVDR) into COS-1 cells. These experiments showed that the flanking region from -298 to -122 directed a 24-fold increase in luciferase activity in response to 1,25-(OH)2D3 provided that the cells were co-transfected with pRSV-hVDR. Within this region, the sequence from position -171 to -123 conferred 1,25-(OH)2D3 responsiveness to both the native P450(24) promoter and the heterologous thymidine kinase promoter. Mutagenesis revealed that the sequence from position -150 to -136 is required for induction by 1,25-(OH)2D3 and that this sequence shares similarity to other vitamin D responsive elements (VDREs) reported for other genes. Gel shift mobility assays showed this region specifically bound a nuclear protein complex from 1,25-(OH)2D3 treated COS-1 cells that had been co-transfected with pRSV-hVDR. The retarded band was specifically competed with the well characterized VDRE from the mouse osteopontin gene. A VDRE at position -150 to -136 in the promoter of the rat P450(24) gene is identified in this study and found to be important in mediating the enhanced expression of the gene by 1,25-(OH)2D3.


Subject(s)
Calcitriol/metabolism , Cytochrome P-450 Enzyme System/genetics , Gene Expression Regulation, Enzymologic , Promoter Regions, Genetic , Steroid Hydroxylases/genetics , Animals , Base Sequence , Cell Line , Cloning, Molecular , DNA , Humans , Mice , Molecular Sequence Data , Rats , Repetitive Sequences, Nucleic Acid , Transfection , Up-Regulation , Vitamin D3 24-Hydroxylase
18.
J Biol Chem ; 269(18): 13437-43, 1994 May 06.
Article in English | MEDLINE | ID: mdl-8175775

ABSTRACT

Elements necessary for the steroid hormone 1 alpha,25-dihydroxyvitamin D3 (1 alpha,25-(OH)2D3) to induce a biological response include the presence of specific intracellular receptors (vitamin D3 receptors (VDR)) and modulation of gene expression via hormone-activated receptor binding to regulatory regions of target genes. These parameters were examined in normal and Epstein-Barr virus-immortalized human B cells and compared with 1 alpha,25-(OH)2D3-responsive cells of the T and monocytic lineages. Although resting tonsillar B cells did not express VDR mRNA, activation of these cells with interleukin-4 induced VDR in the absence of exogenously supplemented 1 alpha,25-(OH)2D3. As indicators of hormone-mediated gene regulation we analyzed modulation of CD23, a common B cell/monocyte surface antigen, and 24-hydroxylase. 1 alpha,25-(OH)2D3 inhibited CD23 expression in U937 cells, yet failed to modulate CD23 expression in B cells. Furthermore, 1 alpha,25-(OH)2D3 induced 24-hydroxylase mRNA expression and metabolic activity in both U937 cells and lectin-activated T cells, yet failed to induce 24-hydroxylase mRNA or its metabolic activity in B cells. These findings suggest that although human B lymphocytes can express VDR mRNA and protein, they exhibit a functional block for vitamin D-dependent gene regulation.


Subject(s)
B-Lymphocytes/metabolism , Calcitriol/physiology , Gene Expression Regulation/physiology , Base Sequence , Cell Line, Transformed , Humans , Molecular Sequence Data , Oligodeoxyribonucleotides , Palatine Tonsil/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Receptors, IgE/genetics
20.
Arch Biochem Biophys ; 293(2): 213-8, 1992 Mar.
Article in English | MEDLINE | ID: mdl-1536558

ABSTRACT

Porcine renodoxon is a kidney mitochondrial iron-sulfur protein (ISP) that functions to transfer electron to cytochromes P450 of the vitamin D pathway. A full-length cDNA clone to porcine renodoxin was isolated in the current investigation and used to study the protein's primary structure and immunological properties. The cysteine ligands for the iron-sulfur center, and the surface protein-binding and phosphorylation sites occupied identical positions in both porcine renodoxin and bovine adrenodoxin. Furthermore, porcine renodoxin was functionally indistinguishable from bovine adrenodoxin and the mature forms of both proteins had the same encoded length and shared approximately 91% sequence similarity. A synthetic peptide to the surface protein-binding region was used to demonstrate the antigenicity of the domain in both the porcine and the bovine ISPs. However, porcine renodoxin displayed only limited immunological identity to other regions of bovine adrenodoxin as measured by competitive enzyme-linked immunosorbent assay. Part of this immunological distinction was attributed to the COOH-terminal processing of porcine renodoxin, an action which negated expression of a COOH-terminal antigenic site that is present in bovine adrenodoxin. Other antigenic differences were linked to charged-residue substitutions that were located in predicted surface domains. The highest frequency of surface-residue substitutions in ferredoxin proteins was predicted for porcine renodoxin, which could provide a basis for understanding why the pig protein appears more antigenically divergent than other ferredoxins.


Subject(s)
Ferredoxins/genetics , Kidney/chemistry , Adrenodoxin/immunology , Amino Acid Sequence , Animals , Antigens, Surface/immunology , Base Sequence , Cattle , Cloning, Molecular , Cross Reactions , DNA/isolation & purification , Enzyme-Linked Immunosorbent Assay , Ferredoxins/immunology , Ferredoxins/isolation & purification , Molecular Sequence Data , Protein Conformation , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...