Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 674
Filter
1.
J Endocrinol Invest ; 43(11): 1623-1630, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32319048

ABSTRACT

INTRODUCTION: Vitamin D is involved in the regulatory mechanisms of ovarian function and is frequently low in PCOS patients. Since obesity and hyperinsulinemic state negatively influenced vitamin D levels, therefore, we evaluated the production of vitamin D at the ovarian level only in lean and normoinsulinemic PCOS subjects. Basal, GnRH analogue-induced ovarian production of 25OH-vitamin D (VitD) and a direct sampling at ovarian vein level were investigated. METHODS: Basal and GnRH analogue-induced hormone levels were evaluated at peripheral level in 45 subjects, aged 18-39 years, and in 22 healthy women with age- and BMI-matched as controls. In 12 PCOS patients, undergoing laparoscopy, a venous sampling at both peripheral and ovarian level was further done. All subjects presented low VitD levels, appropriate to the season and with no difference between PCOS and control subjects. RESULTS: GnRH analogue significantly stimulated plasma LH, FSH, 17-OHP and estradiol secretion (p from < 0.05 to < 0.001 vs basal levels), whereas no effect was observed on both serum AMH and VitD concentrations in all groups. A significant difference (p < 0.006), between peripheral and ovarian veins, was observed in both AMH and estradiol levels in PCOS subjects, while no gradient of VitD was detected. CONCLUSIONS: All patients presented with low VitD levels. The absence of any VitD variation, both at basal and after GnRH analogue administration, or at peripheral-ovarian vein gradient, suggests no pituitary-ovarian axis involvement in VitD production or its direct ovarian production in lean and normoinsulinemic PCOS subjects.


Subject(s)
Hydroxycholecalciferols/metabolism , Ovary/metabolism , Polycystic Ovary Syndrome/metabolism , Adolescent , Adult , Blood Specimen Collection , Case-Control Studies , Female , Follicle Stimulating Hormone/blood , Humans , Luteinizing Hormone/blood , Ovary/blood supply , Ovary/pathology , Polycystic Ovary Syndrome/pathology , Vitamin D/blood , Young Adult
2.
Arch Anim Nutr ; 73(6): 445-456, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31553227

ABSTRACT

To investigate effects of dietary calcium (Ca) and phosphorus (P) levels and 25-hydroxycholecalciferol (25OHD3) supplementation on performance and bone properties of broiler starters, 224 male Arbor Acre broilers were used in a 21-d trial. Broilers were allotted to one of four treatments in a 2 × 2 factorial arrangement including diets either normal or low in Ca and P, which were further supplemented or not with 69 µg 25OHD3/kg feed. Feeding low Ca and P diets significantly reduced performance of boilers and decreased ash, Ca, P and hydroxyproline contents in tibias and femurs (p < 0.05). Ultimate load, bending moment, stiffness and energy to fail were decreased (p < 0.05) in broilers fed diets deficient in Ca and P. Addition of 25OHD3 did not influence performance but significantly increased serum 25OHD3 levels. Furthermore, the addition of 25OHD3 caused an increased tibial and femoral bone density and femoral hydroxyproline content (p < 0.05), increased bending moment in tibias (p < 0.05), and enhanced ultimate load and bending moment in femurs (p < 0.05). No significant interactions were observed for bone properties. Overall, feeding 25OHD3 at 69 µg/kg feed to broilers had no effect on growth performance but partly improved bone biochemical and biomechanical properties of broiler starters.


Subject(s)
Calcium, Dietary/metabolism , Chickens/physiology , Femur/drug effects , Hydroxycholecalciferols/metabolism , Phosphorus, Dietary/metabolism , Tibia/physiology , Animal Feed/analysis , Animals , Biomechanical Phenomena , Chickens/growth & development , Diet/veterinary , Dietary Supplements/analysis , Femur/physiology , Hydroxycholecalciferols/administration & dosage , Male , Random Allocation , Tibia/drug effects
3.
Sci Rep ; 9(1): 12682, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31481665

ABSTRACT

We previously presented the protein-protein interaction network of schizophrenia associated genes, and from it, the drug-protein interactome which showed the drugs that target any of the proteins in the interactome. Here, we studied these drugs further to identify whether any of them may potentially be repurposable for schizophrenia. In schizophrenia, gene expression has been described as a measurable aspect of the disease reflecting the action of risk genes. We studied each of the drugs from the interactome using the BaseSpace Correlation Engine, and shortlisted those that had a negative correlation with differential gene expression of schizophrenia. This analysis resulted in 12 drugs whose differential gene expression (drug versus normal) had an anti-correlation with differential expression for schizophrenia (disorder versus normal). Some of these drugs were already being tested for their clinical activity in schizophrenia and other neuropsychiatric disorders. Several proteins in the protein interactome of the targets of several of these drugs were associated with various neuropsychiatric disorders. The network of genes with opposite drug-induced versus schizophrenia-associated expression profiles were significantly enriched in pathways relevant to schizophrenia etiology and GWAS genes associated with traits or diseases that had a pathophysiological overlap with schizophrenia. Drugs that targeted the same genes as the shortlisted drugs, have also demonstrated clinical activity in schizophrenia and other related disorders. This integrated computational analysis will help translate insights from the schizophrenia drug-protein interactome to clinical research - an important step, especially in the field of psychiatric drug development which faces a high failure rate.


Subject(s)
Anticonvulsants/therapeutic use , Drug Repositioning , Protein Interaction Maps/genetics , Schizophrenia/drug therapy , Acetazolamide/chemistry , Acetazolamide/metabolism , Acetazolamide/therapeutic use , Anticonvulsants/chemistry , Anticonvulsants/metabolism , Carbonic Anhydrases/chemistry , Carbonic Anhydrases/metabolism , Gene Expression Regulation , Genome-Wide Association Study , Humans , Hydroxycholecalciferols/chemistry , Hydroxycholecalciferols/metabolism , Hydroxycholecalciferols/therapeutic use , Receptors, Calcitriol/chemistry , Receptors, Calcitriol/metabolism , Schizophrenia/pathology
4.
Poult Sci ; 97(4): 1263-1270, 2018 Apr 01.
Article in English | MEDLINE | ID: mdl-29452375

ABSTRACT

1α-Hydroxycholecalciferol (1α-OH-D3) is a vitamin D derivative. The objective of this study was to evaluate the effects of 1α-OH-D3 on the growth and the mRNA expression of vitamin D receptor (VDR) in the small intestine and kidney of chickens. A total of 240 males of one-day-old Ross 308 broilers was randomly assigned to 4 treatments with 5 replicates of 12 birds per replicate. Three levels of 1α-OH-D3 (1.25, 2.5, and 5 µg/kg) were added to a basal diet containing 0.50% calcium (Ca), 0.25% non-phytate phosphorus (NPP), and without supplemental cholecalciferol (vitamin D3). The control diet contained 1.00% Ca, 0.45% NPP, and 25 µg/kg cholecalciferol. Dietary 1α-OH-D3 levels linearly improved the average daily feed intake (ADFI), average daily gain (ADG), femur and tibia mineralization, and plasma Ca concentration, and retained Ca and total phosphorus (tP) amounts in broilers from 1 to 21 d of age (P < 0.05). In addition, 1α-OH-D3 also linearly up-regulated the mRNA expression levels of VDR in the duodenum as well as those of VDR and sodium-phosphate cotransporter NaPi-IIa and NaPi-IIc in the kidney of broilers (P < 0.05). However, 1α-OH-D3 did not affect the mRNA levels of 25-hydroxylase in the liver or NaPi-IIb in the duodenum (P > 0.05). No differences were observed in the ADFI, ADG, bone length, plasma mineral concentration, retained tP amount, or the mRNA levels of the above genes (except for VDR in the kidney) between the birds fed the diet with 5 µg/kg 1α-OH-D3 and the birds fed the control diet (P > 0.05). By contrast, the weight, ash weight, ash percentage, and Ca percentage of the bone, retained Ca amount, and the mRNA level of VDR in the kidney were lower in the birds fed the diet with 5 µg/kg 1α-OH-D3 than in the birds fed the control diet (P < 0.05). These data indicate that 1α-OH-D3 up-regulates the gene expression of VDR in the small intestine and kidney at the transcriptional level, thereby improving the growth performance and bone mineralization of broiler chickens from 1 to 21 d of age.


Subject(s)
Avian Proteins/genetics , Chickens/growth & development , Chickens/genetics , Hydroxycholecalciferols/metabolism , Phosphorus, Dietary/metabolism , Receptors, Calcitriol/genetics , Up-Regulation , Animal Feed/analysis , Animals , Avian Proteins/metabolism , Diet/veterinary , Dietary Supplements/analysis , Dose-Response Relationship, Drug , Gene Expression , Hydroxycholecalciferols/administration & dosage , Intestine, Small/metabolism , Kidney/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Random Allocation , Receptors, Calcitriol/metabolism
5.
Clin Biochem ; 50(18): 988-996, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28697996

ABSTRACT

BACKGROUND: Analysis of serum 25-hydroxyvitamin D (s-25(OH)D) may be complicated by the less active or in-active vitamin D metabolite C3-epi-25(OH)D3 (C3-epimer). We aimed to explore the relationship between s-C3-epimer and s-25(OH)D and other determinants and describe the longitudinal course of the C3-epimer fraction in paired mother-child samples. METHOD: S-25(OH)D and s-C3-epimer were estimated by liquid chromatography mass spectrometry in 290 mother-infant pairs from the population-based Odense Child Cohort. Longitudinal analyses were feasible in two subcohorts; B) early and late pregnancy, cord, three and 18months (n=132); and C) early and late pregnancy, delivery and cord (n=105). RESULTS: Mean s-25(OH)D was 50.6-110.4nmol/L at the six time points. The mean C3-epimer fraction was 10.1% at three months, 1.1%-3.0% at the other time points. In multivariate analyses, the s-C3-epimer correlated with s-25(OH)D (all time points, p<0.001), and season, maternal and infant age and maternal vitamin D supplementation at some time points. The C3-epimer fraction fluctuated between adjacent time points. By cosinor analyses, a season-dependent sinusoidal pattern for s-25(OH)D and C3-epimer fraction was found and changes between adjacent time points depended on season (p<0.007 or trend). In early infancy, subtraction of the C3-epi-25(OH)D3 from total s-25(OH)D resulted in reclassification of 8% of the children by use of the 75nmol/L cut off for s-25(OH)D. CONLCUSION: The s-C3-epimer was independently correlated to s-25(OH)D, season, maternal vitamin D supplementation, maternal and infant age. The C3-epimer fraction was only of clinical importance in early infancy, where it could lead to misclassification of the vitamin D status.


Subject(s)
Calcifediol/metabolism , Vitamin D/analogs & derivatives , Calcifediol/physiology , Child , Child, Preschool , Chromatography, Liquid , Cohort Studies , Dietary Supplements , Female , Fetal Blood/metabolism , Humans , Hydroxycholecalciferols/blood , Hydroxycholecalciferols/metabolism , Infant , Male , Pregnancy , Tandem Mass Spectrometry , Vitamin D/blood , Vitamin D/metabolism , Vitamin D Deficiency/blood , Vitamins/blood
6.
J Steroid Biochem Mol Biol ; 173: 42-56, 2017 10.
Article in English | MEDLINE | ID: mdl-27693422

ABSTRACT

The classical pathway of vitamin D activation follows the sequence D3→25(OH)D3→1,25(OH)2D3 with the final product acting on the receptor for vitamin D (VDR). An alternative pathway can be started by the action of CYP11A1 on the side chain of D3, primarily producing 20(OH)D3, 22(OH)D3, 20,23(OH)2D3, 20,22(OH)2D3 and 17,20,23(OH)3D3. Some of these metabolites are hydroxylated by CYP27B1 at C1α, by CYP24A1 at C24 and C25, and by CYP27A1 at C25 and C26. The products of these pathways are biologically active. In the epidermis and/or serum or adrenals we detected 20(OH)D3, 22(OH)D3, 20,22(OH)2D3, 20,23(OH)2D3, 17,20,23(OH)3D3, 1,20(OH)2D3, 1,20,23(OH)3D3, 1,20,22(OH)3D3, 20,24(OH)2D3, 1,20,24(OH)3D3, 20,25(OH)2D3, 1,20,25(OH)3D3, 20,26(OH)2D3 and 1,20,26(OH)3D3. 20(OH)D3 and 20,23(OH)2D3 are non-calcemic, while the addition of an OH at C1α confers some calcemic activity. Molecular modeling and functional assays show that the major products of the pathway can act as "biased" agonists for the VDR with high docking scores to the ligand binding domain (LBD), but lower than that of 1,25(OH)2D3. Importantly, cell based functional receptor studies and molecular modeling have identified the novel secosteroids as inverse agonists of both RORα and RORγ receptors. Specifically, they have high docking scores using crystal structures of RORα and RORγ LBDs. Furthermore, 20(OH)D3 and 20,23(OH)2D3 have been tested in a cell model that expresses a Tet-on RORα or RORγ vector and a RORE-LUC reporter (ROR-responsive element), and in a mammalian 2-hybrid model that test interactions between an LBD-interacting LXXLL-peptide and the LBD of RORα/γ. These assays demonstrated that the novel secosteroids have ROR-antagonist activities that were further confirmed by the inhibition of IL17 promoter activity in cells overexpressing RORα/γ. In conclusion, endogenously produced novel D3 hydroxy-derivatives can act both as "biased" agonists of the VDR and/or inverse agonists of RORα/γ. We suggest that the identification of large number of endogenously produced alternative hydroxy-metabolites of D3 that are biologically active, and of possible alternative receptors, may offer an explanation for the pleiotropic and diverse activities of vitamin D, previously assigned solely to 1,25(OH)2D3 and VDR.


Subject(s)
Hydroxycholecalciferols/metabolism , Hydroxycholecalciferols/pharmacology , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Calcitriol/metabolism , Vitamins/metabolism , Vitamins/pharmacology , Animals , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Humans , Models, Molecular , Nuclear Receptor Subfamily 1, Group F, Member 1/agonists , Nuclear Receptor Subfamily 1, Group F, Member 3/agonists , Receptors, Calcitriol/agonists
7.
J Cell Biochem ; 118(5): 1050-1064, 2017 05.
Article in English | MEDLINE | ID: mdl-27567005

ABSTRACT

Epidemiological and clinical data suggest adverse cardiovascular outcomes with respect to vitamin D deficiency. Here, we explored the effects of vitamin D in atherosclerotic plaque calcification in vivo by utilizing vitamin D receptor (Vdr)-deficient mice in an Apoe-/- background. Animals were fed a high-fat diet (HFD) for either 12 or 18 weeks and then examined for atherosclerotic plaque development. In order to prevent calcium deficiency, Vdr-/- and Apoe-/- ;Vdr-/- animals were fed a high-calcium rescue diet prior to initiation of the HFD feeding and supplemented with high-calcium water during HFD feeding. Although calcium supplementation improved bone mass in Vdr-/- and Apoe-/- ;Vdr-/- mice, neither strain was fully rescued. Systemic inflammatory responses observed in the absence of VDR were exaggerated in Apoe-/- mice. Whereas, hyperlipidemic profiles seen in Apoe-/- mice were ameliorated in the absence of VDR. Micro-computed tomography (µCT) analysis revealed that six out of eight Apoe-/- animals developed atherosclerotic plaque calcification following 12 weeks of HFD feeding and 100% of the mice developed plaque calcification after 18 weeks. In contrast, although atherosclerotic lesions were evident in Apoe-/- ;Vdr-/- mice at 12 and 18 weeks of HFD challenge, none of these animals developed plaque calcification at either time point. The active vitamin D hormone, 1,25(OH)2 D3 likely increased calcification in aortic smooth muscle cells perhaps by directly modulating expression of Alpl, Rankl, and Opg. Our data suggest that the absence of VDR inhibits atherosclerotic plaque calcification in hypercholesterolemic Apoe-/- mice, providing additional insight into the role of vitamin D in atherosclerotic plaque calcification. J. Cell. Biochem. 118: 1050-1064, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Apolipoproteins E/deficiency , Calcium/administration & dosage , Hypercholesterolemia/complications , Plaque, Atherosclerotic/prevention & control , Receptors, Calcitriol/deficiency , Animals , Apolipoproteins E/genetics , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Hydroxycholecalciferols/metabolism , Hypercholesterolemia/chemically induced , Mice , Mice, Knockout , Plaque, Atherosclerotic/diagnostic imaging , Plaque, Atherosclerotic/genetics , Receptors, Calcitriol/genetics , X-Ray Microtomography
8.
Poult Sci ; 95(3): 570-80, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26666254

ABSTRACT

While it is well established that active vitamin D treatment increases dietary phytate phosphate utilization, the mechanism by which intestinal alkaline phosphatase (IAP) participates in phytate phosphate use is less clear. The ability of human IAP (hIAP) oral antibodies to prevent dietary phytate phosphate utilization in the presence of 1α-hydroxycholecalciferol (1α-(OH) D3) in a chick model was investigated. hIAP specific chicken immunoglobulin Y (IgY) antibodies were generated by inoculating laying hens with 17 synthetic peptides derived from the human IAP amino acid sequence and harvesting egg yolk. Western blot analysis showed all antibodies recognized hIAP and 6 of the 8 antibodies selected showed modest inhibition of hIAP activity in vitro (6 to 33% inhibition). In chicks where dietary phosphate was primarily in the form of phytate, 4 selected hIAP antibodies inhibited 1α-(OH) D3-induced increases in blood phosphate, one of which, generated against selected peptide (MFPMGTPD), was as effective as sevelamer hydrochloride in preventing the 1α-(OH) D3-induced increase in blood phosphate, but ineffective in preventing an increase in body weight gain and bone ash induced by 1α-(OH) D3. These studies demonstrated that orally-delivered antibodies to IAP limit dietary phytate-phosphate utilization in chicks treated with 1α-(OH) D3, and implicate IAP as an important host enzyme in increasing phytate phosphate bioavailability in 1α-(OH) D3 fed chicks.


Subject(s)
Alkaline Phosphatase/metabolism , Antibodies/metabolism , Chickens/metabolism , Diet/veterinary , Hydroxycholecalciferols/metabolism , Phosphates/metabolism , Animals , Biological Availability , GPI-Linked Proteins/metabolism , Male , Phosphates/blood , Phytic Acid/metabolism
9.
PLoS One ; 10(10): e0140478, 2015.
Article in English | MEDLINE | ID: mdl-26488576

ABSTRACT

Genetic polymorphisms in vitamin D metabolism and signaling genes have been inconsistently associated with risk of breast cancer, though few studies have examined SNPs in vitamin D-related genes other than the vitamin D receptor (VDR) gene and particularly have not examined the association with the retinoid X receptor alpha (RXRA) gene which may be a key vitamin D pathway gene. We conducted a nested case-control study of 734 cases and 1435 individually matched controls from a population-based prospective cohort study, the Northern Sweden Mammary Screening Cohort. Tag and functional SNPs were genotyped for the VDR, cytochrome p450 24A1 (CYP24A1), and RXRA genes. We also genotyped specific SNPs in four other genes related to vitamin D metabolism and signaling (GC/VDBP, CYP2R1, DHCR7, and CYP27B1). SNPs in the CYP2R1, DHCR7, and VDBP gene regions that were associated with circulating 25(OH)D concentration in GWAS were also associated with plasma 25(OH)D in our study (p-trend <0.005). After taking into account the false discovery rate, these SNPs were not significantly associated with breast cancer risk, nor were any of the other SNPs or haplotypes in VDR, RXRA, and CYP24A1. We observed no statistically significant associations between polymorphisms or haplotypes in key vitamin D-related genes and risk of breast cancer. These results, combined with the observation in this cohort and most other prospective studies of no association of circulating 25(OH)D with breast cancer risk, do not support an association between vitamin D and breast cancer risk.


Subject(s)
Breast Neoplasms/genetics , Hydroxycholecalciferols/blood , Receptors, Calcitriol/genetics , Retinoid X Receptor alpha/genetics , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , Aged , Breast Neoplasms/blood , Case-Control Studies , Cholestanetriol 26-Monooxygenase/genetics , Cohort Studies , Cytochrome P450 Family 2 , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Genotype , Humans , Hydroxycholecalciferols/metabolism , Middle Aged , Oxidoreductases Acting on CH-CH Group Donors/genetics , Polymorphism, Single Nucleotide , Prospective Studies , Signal Transduction/genetics , Sweden , Vitamin D-Binding Protein/genetics , Vitamin D3 24-Hydroxylase/genetics
10.
J Clin Endocrinol Metab ; 100(11): 4140-5, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26312580

ABSTRACT

CONTEXT: It has been proposed that serum free 25-hydroxyvitamin D [25(OH)D] may better reflect vitamin D action than total 25(OH)D. An ELISA for serum free 25(OH)D has recently become available, permitting direct assay. OBJECTIVE: To determine whether serum free 25(OH)D provides additional information in relation to calcium absorption and other biomarkers of vitamin D action compared to total serum 25(OH)D. SETTING: Ambulatory research setting in a teaching hospital. OUTCOME: Serum free 25(OH)D measured in a previously performed study of varied doses of vitamin D3 (placebo and 800, 2000, and 4000 IU) on calcium absorption, PTH, procollagen type 1 N-terminal propeptide, and C-terminal telopeptides of type I collagen. Free 25(OH)D was measured by ELISA. Calcium absorption was measured at baseline and at 10 weeks using stable dual calcium isotopes. RESULTS: Seventy-one subjects completed this randomized, placebo-controlled trial. Baseline group mean free and total 25(OH)D varied from 4.7 ± 1.8 to 5.4 ± 1.5 pg/mL, and from 23.7 ± 5.9 to 25.9 ± 6.1 ng/mL, respectively. Participants assigned to the 4000-IU dose arm achieved free 25(OH)D levels of 10.4 pg/mL and total 25(OH)D levels of 40.4 ng/mL. Total and free 25(OH)D were highly correlated at baseline and after increasing vitamin D dosing (r = 0.80 and 0.85, respectively). Free 25(OH)D closely reflected changes in total 25(OH)D. PTH was similarly correlated at baseline and follow-up with total and free 25(OH)D. Serum C-terminal telopeptides of type I collagen had a moderate positive correlation with total and free 25(OH)D at follow-up. The serum 1,25-dihydroxyvitamin D change increased significantly with the change in 25(OH)D but not with the change in free 25(OH)D. CONCLUSION: There was no advantage from measuring free over total 25(OH)D in assessing the response of calcium absorption, PTH, and markers of bone turnover to vitamin D. Free 25(OH)D responded to increasing doses of vitamin D in a similar fashion to total 25(OH)D.


Subject(s)
Bone Remodeling , Bone and Bones/metabolism , Calcium/metabolism , Hydroxycholecalciferols/blood , Parathyroid Hormone/metabolism , Vitamins/blood , Adult , Aged , Biomarkers , Collagen Type I/blood , Dose-Response Relationship, Drug , Female , Humans , Hydroxycholecalciferols/metabolism , Hydroxycholecalciferols/pharmacology , Male , Middle Aged , Vitamins/pharmacology , Young Adult
11.
Mar Drugs ; 13(2): 741-55, 2015 Jan 28.
Article in English | MEDLINE | ID: mdl-25636157

ABSTRACT

The high incidence of cardiovascular disease and vitamin D deficiency in chronic kidney disease patients is well known. Vitamin D activation by omega-3 fatty acid (FA) supplementation may explain the cardioprotective effects exerted by omega-3 FA. We hypothesized that omega-3 FA and 25-hydroxyvitamin D (25(OH)D) supplementation may increase 1,25-dihydroxyvitamin D (1,25(OH)2D) levels compared to 25(OH)D supplementation alone in hemodialysis (HD) patients that have insufficient or deficient 25(OH)D levels. We enrolled patients that were treated for at least six months with 25(OH)D < 30 ng/mL (NCT01596842). Patients were randomized to treatment for 12 weeks with cholecalciferol supplemented with omega-3 FA or a placebo. Levels of 25(OH)D and 1,25(OH)2D were measured after 12 weeks. The erythrocyte membrane FA contents were also measured. Levels of 25(OH)D were increased in both groups at 12 weeks compared to baseline. The 1,25(OH)2D levels at 12 weeks compared to baseline showed a tendency to increase in the omega-3 FA group. The oleic acid and monounsaturated FA content decreased, while the omega-3 index increased in the omega-3 FA group. Omega-3 FA supplementation may be partly associated with vitamin D activation, although increased 25(OH)D levels caused by short-term cholecalciferol supplementation were not associated with vitamin D activation in HD patients.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Renal Dialysis/adverse effects , Vitamin D/metabolism , Activation, Metabolic/drug effects , Aged , Cholecalciferol/pharmacology , Diet , Dietary Supplements , Double-Blind Method , Erythrocyte Membrane/drug effects , Erythrocyte Membrane/metabolism , Female , Humans , Hydroxycholecalciferols/metabolism , Male , Middle Aged , Oleic Acid/blood , Pilot Projects , Vitamin D Deficiency/drug therapy , Vitamin D Deficiency/etiology , Vitamins/pharmacology
12.
Curr Vasc Pharmacol ; 13(3): 392-8, 2015.
Article in English | MEDLINE | ID: mdl-24329737

ABSTRACT

Levels of 25-hydroxy vitamin D [25(OH)D] are reported to be decreased in cardiovascular disease (CVD) and in other chronic immunopathologies. Vitamin D (vitD) has been shown to be significantly linked to mortality, and is thought to be a predictor of survival. Therefore, supplementation with vitD has been suggested as an option to improve clinical outcomes. In contrast to the causal assumption, we hypothesize that the decreased vitD levels, seen in patients with CVD and chronic immunopathologies is secondary to inflammation and not as pathophysiologically relevant as currently suggested. Under these conditions, low vitD might be mainly caused by oxidative stress that results from chronic, immune-mediated vascular and systemic inflammation seen in patients with CVD. The oxidative environment most likely causes biodegradation of vitD and interferes with key enzymes, disturbing the biosynthesis of 25(OH)D and 1,25(OH)D. Thus far, no clear evidence of a beneficial effect of vitD supplements exists, beyond treating vitD deficiency to improve skeletal health. Moreover, a prolonged and/or high dose vitD supplementation, unless needed to correct actual vitD deficiency [levels of 25(OH)D<20 ng/ml)] may even be immunologically harmful by downregulating Th1 immune responses and indirectly upregulating Th2 immune activation with potential detrimental metabolic and cardiovascular effects. Large randomized controlled studies of vitD with multiple outcomes (skeletal, metabolic, cardiovascular and mental) are urgently needed.


Subject(s)
Atherosclerosis/drug therapy , Atherosclerosis/pathology , Inflammation/drug therapy , Inflammation/pathology , Vitamin D/physiology , Vitamin D/therapeutic use , Animals , Humans , Hydroxycholecalciferols/metabolism , Vitamin D/metabolism
13.
FEBS J ; 281(14): 3280-96, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24893882

ABSTRACT

CYP24A1 is the multicatalytic cytochrome P450 responsible for the catabolism of vitamin D via the C23- and C24-oxidation pathways. We successfully expressed the labile human enzyme in Escherichia coli and partially purified it in an active state that permitted detailed characterization of its metabolism of 1,25-dihydroxyvitamin D3 [1,25(OH)2 D3] and the intermediates of the C24-oxidation pathway in a phospholipid-vesicle reconstituted system. The C24-oxidation pathway intermediates, 1,24,25-trihydroxyvitamin D3, 24-oxo-1,25-dihydroxyvitamin D3, 24-oxo-1,23,25-trihydroxyvitamin D3 and tetranor-1,23-dihydroxyvitamin D3, were enzymatically produced from 1,25(OH)2 D3 using rat CYP24A1. Both 1,25(OH)2 D3 and 1,23-dihydroxy-24,25,26,27-tetranorvitamin D3 were found to partition strongly into the phospholipid bilayer when in aqueous medium. Changes to the phospholipid concentration did not affect the kinetic parameters for the metabolism of 1,25(OH)2 D3 by CYP24A1, indicating that it is the concentration of substrates in the membrane phase (mol substrate·mol phospholipid(-1) ) that determines their rate of metabolism. CYP24A1 exhibited Km values for the different C24-intermediates ranging from 0.34 to 15 mmol·mol phospholipid(-1) , with 24-oxo-1,23,25-trihydroxyvitamin D3 [24-oxo-1,23,25(OH)3 D3] displaying the lowest and 1,24,25-trihydroxyvitamin D3 [1,24,25(OH)3 D3] displaying the highest. The kcat values varied by up to 3.8-fold, with 1,24,25(OH)3 D3 displaying the highest kcat (34 min(-1) ) and 24-oxo-1,23,25(OH)3 D3 the lowest. The data show that the cleavage of the side chain of 24-oxo-1,23,25(OH)3 D3 occurs with the highest catalytic efficiency (kcat /Km ) and produces 1-hydroxy-23-oxo-24,25,26,27-tetranorvitamin D3 and not 1,23-dihydroxy-24,25,26,27-tetranorvitamin D3, as the primary product. These kinetic analyses also show that intermediates of the C24-oxidation pathway effectively compete with precursor substrates for binding to the active site of the enzyme, which manifests as an accumulation of intermediates, indicating that they dissociate after each catalytic step.


Subject(s)
Calcitriol/metabolism , Steroid Hydroxylases/metabolism , Adrenodoxin/metabolism , Animals , Calcitriol/analogs & derivatives , Humans , Hydroxycholecalciferols/metabolism , Kinetics , Mitochondrial Membranes/metabolism , Rats , Steroid Hydroxylases/isolation & purification , Vitamin D3 24-Hydroxylase
14.
Chem Pharm Bull (Tokyo) ; 62(2): 182-4, 2014.
Article in English | MEDLINE | ID: mdl-24492588

ABSTRACT

Our previous studies revealed that recombinant human CYP3A4 converted 2α-(3-hydroxypropoxy)-1α,25-dihydroxyvitamin D3 (O2C3), which was a more potent binder to vitamin D receptor (VDR) than the natural hormone, 1α,25-dihydroxyvitamin D3 (1α,25(OH)2D3, 1), to 1α,2α,25-trihydroxyvitamin D3 (2). Here, we synthesized 2 using the Trost Pd-mediated coupling reaction between an A-ring precursor and a CD-ring bromoolefin and evaluated its preliminary biological activity. We found that metabolite 2 from O2C3 was still active as a VDR ligand while maintaining human VDR binding affinity (27.3% of 1α,25(OH)2D3) and HL-60 cell differentiation activity (62% of 1α,25(OH)2D3).


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Hydroxycholecalciferols/metabolism , Hydroxycholecalciferols/pharmacology , Vitamin D/analogs & derivatives , Vitamins/metabolism , Vitamins/pharmacology , Cell Differentiation/drug effects , HL-60 Cells , Humans , Hydroxycholecalciferols/chemistry , Protein Binding , Receptors, Calcitriol/metabolism , Vitamin D/chemistry , Vitamin D/metabolism , Vitamin D/pharmacology , Vitamins/chemistry
15.
Nat Prod Res ; 28(7): 444-8, 2014.
Article in English | MEDLINE | ID: mdl-24456427

ABSTRACT

The microbial transformation of vitamin D3 (1) by the fungi Candida maltosa R42 and Botrytis allii NRRL 2502 was investigated. Incubation of compound 1 with C. maltosa R42 and B. allii NRRL 2502 produced the same three more polar metabolites in small yields. The main metabolite was identified as 1α-hydroxyvitamin D3 (2). This biotransformation has utility as a possible tool for the production of 1α-hydroxyvitamin D3 from the readily available vitamin D3 for patients with compromised kidney function.


Subject(s)
Botrytis/metabolism , Candida/metabolism , Cholecalciferol/metabolism , Hydroxycholecalciferols/metabolism , Cholecalciferol/chemistry , Humans , Hydroxycholecalciferols/chemistry , Molecular Structure
16.
Psychopharmacology (Berl) ; 231(17): 3445-51, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24448902

ABSTRACT

RATIONALE: Chronic stress or hypercortisolism may increase the risks of depression, cardiac disorders, and osteoporosis, which are also associated with vitamin D (VD) deficiency. Both glucocorticoid receptor (GR) and vitamin D receptor (VDR) are widely distributed and affect many aspects of human physiology. The cross talk between the two steroids is pervasive, but the effect of glucocorticoids on circulating VD and local VD metabolism remains elusive. OBJECTIVES: To fill this critical gap, we assessed the alterations of circulating VD and VD intracrine system in the brain and myocardium of rats treated with two different doses (0.2 and 2 mg/kg/day, respectively) of dexamethasone (Dex). RESULTS: Daily treatment with 2 mg/kg of Dex for 10 days induced the rats to a depressive-like state and decreased the expression of both VDR and the cytochromes P450 enzymes involved in VD activation (CYP27B1) and catabolism (CYP24A1) in the prefrontal cortex and hippocampus. Meanwhile, the dose of 0.2 mg/kg Dex increased the expression of VDR in the prefrontal cortex but inhibited CYP27B1/CYP24A1/VDR expression in the hippocampus. Similarly, in the myocardium, the rats treated with Dex showed significantly lower expression of CYP27B1/CYP24A1/VDR. Renal VD metabolism and serum VD status were unchanged in 0.2 mg/kg Dex-treated rats. However, the higher dose suppressed the three key players involved in VD metabolism but did not alter serum VD levels. CONCLUSION: These data provide new evidence that glucocorticoids could affect intracrine actions of VD in the brain and myocardium, which suggests the potential involvement of VD in the neural and cardiac dysfunctions induced by glucocorticoid excess.


Subject(s)
Brain Chemistry/drug effects , Dexamethasone/pharmacology , Heart/drug effects , Myocardium/metabolism , Vitamin D/metabolism , Vitamins/metabolism , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/biosynthesis , Anhedonia/drug effects , Animals , Behavior, Animal/drug effects , Body Weight/drug effects , Dihydroxycholecalciferols/metabolism , Hydroxycholecalciferols/metabolism , Kidney/drug effects , Kidney/metabolism , Male , Rats , Rats, Sprague-Dawley , Swimming/psychology , Vitamin D3 24-Hydroxylase/biosynthesis
17.
J Cell Biochem ; 114(10): 2293-305, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23606409

ABSTRACT

3-epi-1α,25-dihydroxyvitamin D3 (3-epi-1α,25(OH)2D3), a natural metabolite of 1α,25-dihydroxyvitamin D3 (1α,25(OH)2D3), exhibits potent vitamin D receptor (VDR)-mediated actions such as inhibition of keratinocyte growth or suppression of parathyroid hormone secretion. These VDR-mediated actions of 3-epi-1α,25(OH)2D3 needed an explanation as 3-epi-1α,25(OH)2D3, unlike 1α,25(OH)2D3, exhibits low affinity towards VDR. Metabolic stability of 3-epi-1α,25(OH)2D3 over 1α,25(OH)2D3 has been hypothesized as a possible explanation. To provide further support for this hypothesis, we now performed comparative metabolism studies between 3-epi-1α,25(OH)2D3 and 1α,25(OH)2D3 using both the technique of isolated rat kidney perfusion and purified rat CYP24A1 in a cell-free reconstituted system. For the first time, these studies resulted in the isolation and identification of 3-epi-calcitroic acid as the final inactive metabolite of 3-epi-1α,25(OH)2D3 produced by rat CYP24A1. Furthermore, under identical experimental conditions, it was noted that the amount of 3-epi-calcitroic acid produced from 3-epi-1α,25(OH)2D3 is threefold less than that of calcitroic acid, the analogous final inactive metabolite produced from 1α,25(OH)2D3 . This key observation finally led us to conclude that the rate of overall side-chain oxidation of 3-epi-1α,25(OH)2D3 by rat CYP24A1 leading to its final inactivation is slower than that of 1α,25(OH)2D3. To elucidate the mechanism responsible for this important finding, we performed a molecular docking analysis using the crystal structure of rat CYP24A1. Docking results suggest that 3-epi-1α,25(OH)2D3, unlike 1α,25(OH)2D3, binds to CYP24A1 in an alternate configuration that destabilizes the formation of the enzyme-substrate complex sufficiently to slow the rate at which 3-epi-1α,25(OH)2D3 is inactivated by CYP24A1 through its metabolism into 3-epi-calcitroic acid.


Subject(s)
Hydroxycholecalciferols/metabolism , Molecular Dynamics Simulation , Steroid Hydroxylases/metabolism , Vitamin D/analogs & derivatives , Animals , Chromatography, High Pressure Liquid , Gas Chromatography-Mass Spectrometry , Rats , Vitamin D/metabolism , Vitamin D3 24-Hydroxylase
18.
Biochimie ; 95(3): 455-60, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22732193

ABSTRACT

The oxysterol 25-hydroxycholesterol is a widely used compound displaying an array of pharmacological actions in in vitro systems and cell based experimental systems. In spite of the frequent use of this compound over the last few decades and a large number of studies in vitro and in vivo, its mechanism of formation in vivo is still not well understood. Cholesterol autoxidation does not seem to be an important contributor to in vivo formation of 25-hydroxycholesterol. A number of different cytochrome P450 enzymes such as CYP27A1 and CYP3A4 have been reported to catalyze the conversion of cholesterol to 25-hydroxycholesterol in vitro, but the importance of these reactions in vivo remains unclear. The dioxygenase enzyme cholesterol 25-hydroxylase has been shown to generate 25-hydroxycholesterol, but in cholesterol 25-hydroxylase knockout mice there are still significant levels of 25-hydroxycholesterol in several tissues. This suggests that cholesterol 25-hydroxylase is not the sole producer of 25-hydroxycholesterol. The relative importance of different mechanisms of formation of 25-hydroxycholesterol in vivo have still to be elucidated. The maintenance of cholesterol homeostasis is of great importance to supply tissues with the appropriate amount of cholesterol and prevent accumulation that may affect health. Numerous articles mention 25-hydroxycholesterol as an important regulator of cholesterol metabolism. However, mice with a disruption of the cholesterol 25-hydroxylase gene regulate cholesterol metabolism normally and patients with highly elevated levels of 25-hydroxycholesterol also display normal cholesterol and bile acid levels. These reports challenge the hypothesis that 25-hydroxycholesterol is an important regulator of cholesterol metabolism. Recent reports suggest that 25-hydroxycholesterol and one of its metabolites may have functions in regulation of humoral immunity. Thus, 25-hydroxycholesterol may be more important as a regulator of immunity than as a regulator of cholesterol metabolism.


Subject(s)
Hydroxycholesterols/metabolism , Alzheimer Disease/enzymology , Alzheimer Disease/genetics , Animals , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Humans , Hydroxycholecalciferols/metabolism , Hydroxycholesterols/immunology , Monocytes/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism
19.
Chem Pharm Bull (Tokyo) ; 60(10): 1343-6, 2012.
Article in English | MEDLINE | ID: mdl-23036975

ABSTRACT

A previous report has demonstrated the existence of a C4-hydroxylated vitamin D(2) metabolite in serum of rats treated with pharmacological doses of vitamin D(2). However, the biological significance and metabolic fate of this metabolite have not been described. To explore its potential biological activities, we therefore synthesized 1α,4α,25-trihydroxyvitamin D(3) and its diastereoisomer, 1α,4ß,25-trihydroxyvitamin D(3), using Trost Pd-mediated coupling reaction, and studied their vitamin D receptor (VDR) binding affinity, osteocalcin promoter transactivation activity, and their further metabolism by human CYP24A1 as well as by human liver microsomal fraction based on CYP- and UDP-glucuronosyltransferases (UGTs)-reactions.


Subject(s)
Glucuronosyltransferase/metabolism , Hydroxycholecalciferols/chemistry , Hydroxycholecalciferols/pharmacology , Steroid Hydroxylases/metabolism , Cell Line , Humans , Hydroxycholecalciferols/chemical synthesis , Hydroxycholecalciferols/metabolism , Microsomes, Liver/metabolism , Osteocalcin/genetics , Receptors, Calcitriol/metabolism , Transcriptional Activation/drug effects , Vitamin D3 24-Hydroxylase
20.
Drug Metab Dispos ; 39(9): 1577-88, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21677063

ABSTRACT

Cytochrome P450scc (CYP11A1) can hydroxylate vitamin D(3), producing 20S-hydroxyvitamin D(3) [20(OH)D(3)] and 20S,23-dihydroxyvitamin D(3) [20,23(OH)(2)D(3)] as the major metabolites. These are biologically active, acting as partial vitamin D receptor (VDR) agonists. Minor products include 17-hydroxyvitamin D(3), 17,20-dihydroxyvitamin D(3), and 17,20,23-trihydroxyvitamin D(3). In the current study, we have further analyzed the reaction products from cytochrome P450scc (P450scc) action on vitamin D(3) and have identified two 22-hydroxy derivatives as products, 22-hydroxyvitamin D(3) [22(OH)D(3)] and 20S,22-dihydroxyvitamin D(3) [20,22(OH)(2)D(3)]. The structures of both of these derivatives were determined by NMR. P450scc could convert purified 22(OH)D(3) to 20,22(OH)(2)D(3). The 20,22(OH)(2)D(3) could also be produced from 20(OH)D(3) and was metabolized to a trihydroxyvitamin D(3) product. We compared the biological activities of these new derivatives with those of 20(OH)D(3), 20,23(OH)(2)D(3), and 1α,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3)]. 1,25(OH)(2)D(3), 20(OH)D(3), 22(OH)D(3), 20,23(OH)(2)D(3), and 20,22(OH)(2)D(3) significantly inhibited keratinocyte proliferation in a dose-dependent manner. The strongest inducers of involucrin expression (a marker of keratinocyte differentiation) were 20,23(OH)(2)D(3), 20,22(OH)(2)D(3), 20(OH)D(3), and 1,25(OH)(2)D(3), with 22(OH)D(3) having a heterogeneous effect. Little or no stimulation of CYP24 mRNA expression was observed for all the analogs tested except for 1,25(OH)(2)D(3). All the compounds stimulated VDR translocation from the cytoplasm to the nucleus with 22(OH)D(3) and 20,22(OH)(2)D(3) having less effect than 1,25(OH)(2)D(3) and 20(OH)D(3). Thus, we have identified 22(OH)D(3) and 20,22(OH)(2)D(3) as products of CYP11A1 action on vitamin D(3) and shown that, like 20(OH)D(3) and 20,23(OH)(2)D(3), they are active on keratinocytes via the VDR, however, showing a degree of phenotypic heterogeneity in comparison with other P450scc-derived hydroxy metabolites of vitamin D(3).


Subject(s)
Calcifediol/analogs & derivatives , Cholecalciferol/metabolism , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Hydroxycholecalciferols/chemistry , Hydroxycholecalciferols/metabolism , Skin/metabolism , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Calcifediol/chemistry , Calcifediol/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Cholecalciferol/chemistry , Dihydroxycholecalciferols/chemistry , Dihydroxycholecalciferols/metabolism , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Magnetic Resonance Spectroscopy/methods , Protein Precursors/genetics , Protein Precursors/metabolism , Protein Transport/drug effects , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Skin/cytology , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Vitamin D3 24-Hydroxylase
SELECTION OF CITATIONS
SEARCH DETAIL
...