Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Bone ; 32(4): 332-40, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12689675

ABSTRACT

Mutations in the 25-hydroxyvitamin D-1alpha-hydroxylase gene (CYP27B1; 1alpha-OHase) cause pseudo vitamin D deficiency rickets (PDDR), while mutations in the vitamin D receptor (VDR) cause hereditary vitamin D resistance rickets. Animal models of both diseases have been engineered. The bone phenotype of VDR-ablated mice can be completely rescued by feeding the animals with a high-calcium, high-phosphorus, high-lactose diet. We have attempted to rescue the PDDR phenotype of mice deficient for the 1alpha-OHase gene by feeding them with the high-calcium diet. The rescue regimen consisted of feeding a diet containing 2% calcium, 1.25% phosphorus, 20% lactose (rescue diet) from 3 weeks of age until sacrifice at 8.5 weeks of age. Blood biochemistry analysis revealed that the rescue diet corrected the hypocalcemia and secondary hyperparathyroidism. Despite the restoration of normocalcemia, 1alpha-OHase(-/-) (and 1alpha-OHase(+/-)) animals fed the rescue diet initially gained weight less rapidly than control mice fed normal mouse chow. Although 1alpha-OHase(-/-) mice fed the rescue diet eventually reached the same weight as control animals, the treatment did not entirely correct bone growth, as femur size remained significantly smaller than that of control. Bone histology and histomorphometry confirmed that the rickets and osteomalacia were cured. The rescue diet also restored the biomechanical properties of the bone tissue within normal parameters. These results demonstrate that correction of the abnormal mineral ion homeostasis by feeding with a high-calcium rescue diet is effective to rescue the PDDR phenotype of 1alpha-OHase mutant mice. This treatment, however, does not appear as effective as 1,25(OH)(2)D(3) replacement therapy since bone growth remained impaired.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/deficiency , Bone and Bones/pathology , Rickets/diet therapy , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , Animals , Bone and Bones/physiopathology , Calcium, Dietary/therapeutic use , Diet , Homeostasis/physiology , Hyperparathyroidism/diet therapy , Hyperparathyroidism/etiology , Hypocalcemia/diet therapy , Hypocalcemia/etiology , Lactose/administration & dosage , Male , Mice , Models, Animal , Phosphorus/administration & dosage , Rickets/complications , Vitamin D Deficiency/enzymology , Vitamin D Deficiency/genetics
2.
Cell ; 107(4): 513-23, 2001 Nov 16.
Article in English | MEDLINE | ID: mdl-11719191

ABSTRACT

In humans, low peak bone mass is a significant risk factor for osteoporosis. We report that LRP5, encoding the low-density lipoprotein receptor-related protein 5, affects bone mass accrual during growth. Mutations in LRP5 cause the autosomal recessive disorder osteoporosis-pseudoglioma syndrome (OPPG). We find that OPPG carriers have reduced bone mass when compared to age- and gender-matched controls. We demonstrate LRP5 expression by osteoblasts in situ and show that LRP5 can transduce Wnt signaling in vitro via the canonical pathway. We further show that a mutant-secreted form of LRP5 can reduce bone thickness in mouse calvarial explant cultures. These data indicate that Wnt-mediated signaling via LRP5 affects bone accrual during growth and is important for the establishment of peak bone mass.


Subject(s)
Bone Density/genetics , Eye Abnormalities/genetics , Eye/embryology , Osteoblasts/metabolism , Osteoporosis/genetics , Receptors, LDL/physiology , Transforming Growth Factor beta , Zebrafish Proteins , Adaptor Proteins, Signal Transducing , Adult , Animals , Animals, Outbred Strains , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/pharmacology , COS Cells , Child , Child, Preschool , Chlorocebus aethiops , Chromosomes, Human, Pair 11/genetics , Culture Media, Conditioned/pharmacology , DNA, Complementary/genetics , Dishevelled Proteins , Female , Genes, Recessive , Heterozygote , Humans , LDL-Receptor Related Proteins , Low Density Lipoprotein Receptor-Related Protein-5 , Male , Mesoderm/cytology , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Phosphoproteins/genetics , Phosphoproteins/physiology , Proteins/genetics , Proteins/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Receptors, LDL/deficiency , Receptors, LDL/genetics , Recombinant Fusion Proteins/physiology , Recombinant Proteins , Signal Transduction , Skull/cytology , Species Specificity , Stromal Cells/cytology , Stromal Cells/drug effects , Syndrome , Transfection , Wnt Proteins , Wnt-5a Protein , Wnt2 Protein , Wnt3 Protein , Wnt4 Protein
3.
Am J Clin Nutr ; 71(5 Suppl): 1317S-24S, 2000 05.
Article in English | MEDLINE | ID: mdl-10799409

ABSTRACT

During pregnancy, maternal serum concentrations of 25-hydroxyvitamin D, the circulating form of vitamin D, correlate with dietary vitamin D intake. Maternal serum concentrations of 1,25-dihydroxyvitamin D, the hormonal circulating and active form of vitamin D, are elevated during pregnancy; 1,25-dihydroxyvitamin D is synthesized mainly by the decidual cells of the placenta and allows for increased calcium absorption. The fetus is entirely dependent on the mother for its supply of 25-hydroxyvitamin D, which is believed to cross the placenta. Hypocalcemia and increased parathyroid hormone secretion induce synthesis of 1,25-dihydroxyvitamin D after birth in both full-term and preterm neonates. Nevertheless, serum concentrations of 25-hydroxyvitamin D are a rate-limiting factor in the synthesis of 1,25-dihydroxyvitamin D. In vitamin D-replete infants, circulating 1,25-dihydroxyvitamin D concentrations are higher than those observed in older infants. In countries where dairy products are not routinely supplemented with vitamin D, maternal vitamin D supplementation during pregnancy is necessary. However, there is no indication for the use of pharmacologic doses of vitamin D or its metabolites in the perinatal period.


Subject(s)
Dietary Supplements , Infant, Newborn/metabolism , Perinatal Care , Pregnancy/metabolism , Vitamin D/metabolism , Female , Fetus/metabolism , Humans , Infant, Premature/metabolism , Nutritional Physiological Phenomena
4.
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
5.
Hum Mol Genet ; 6(4): 539-49, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9097956

ABSTRACT

Mutations in the PEX gene at Xp22.1 (phosphate-regulating gene with homologies to endopeptidases, on the X-chromosome), are responsible for X-linked hypophosphataemic rickets (HYP). Homology of PEX to the M13 family of Zn2+ metallopeptidases which include neprilysin (NEP) as prototype, has raised important questions regarding PEX function at the molecular level. The aim of this study was to analyse 99 HYP families for PEX gene mutations, and to correlate predicted changes in the protein structure with Zn2+ metallopeptidase gene function. Primers flanking 22 characterised exons were used to amplify DNA by PCR, and SSCP was then used to screen for mutations. Deletions, insertions, nonsense mutations, stop codons and splice mutations occurred in 83% of families screened for in all 22 exons, and 51% of a separate set of families screened in 17 PEX gene exons. Missense mutations in four regions of the gene were informative regarding function, with one mutation in the Zn2+-binding site predicted to alter substrate enzyme interaction and catalysis. Computer analysis of the remaining mutations predicted changes in secondary structure, N-glycosylation, protein phosphorylation and catalytic site molecular structure. The wide range of mutations that align with regions required for protease activity in NEP suggests that PEX also functions as a protease, and may act by processing factor(s) involved in bone mineral metabolism.


Subject(s)
Hypophosphatemia, Familial/genetics , Mutation , Proteins/genetics , Amino Acid Sequence , Base Sequence , Binding Sites , Cloning, Molecular , Codon, Terminator , DNA Primers , DNA, Complementary/chemistry , Databases, Factual , Humans , Metalloendopeptidases/chemistry , Metalloendopeptidases/genetics , Molecular Sequence Data , PHEX Phosphate Regulating Neutral Endopeptidase , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Proteins/chemistry , Proteins/metabolism , RNA Splicing , Sequence Deletion , Sequence Homology, Amino Acid
6.
Genomics ; 36(1): 22-8, 1996 Aug 15.
Article in English | MEDLINE | ID: mdl-8812412

ABSTRACT

The recently identified human PEX gene apparently encodes for a neutral endopeptidase that is mutated in patients with X-linked hypophosphatemia. The 3' and 5' ends of the coding region of PEX have not been cloned, nor has the tissue expression of the gene been identified. Here we report the isolation and characterization of the complete open reading frame of the mouse Pex gene and the demonstration of its expression in bone. Mouse Pex cDNA is predicted to encode a protein of 749 amino acids with 95% identity to the available human PEX sequence and significant homology to members of the membrane-bound metalloendopeptidase family. Northern blot analysis revealed a 6.6-kb transcript in bone and in cultured osteoblasts from normal mice that was not detectable in samples from the Hyp mouse, the murine homolog of human X-linked hypophosphatemia. Pex transcripts were, however, detectable in Hyp bone by RT-PCR amplification. Of particular interest, a cDNA clone from rat incisor shows 93% sequence identity to the 5' end of Pex cDNA, suggesting that Pex may be expressed in another calcified tissue, the tooth. The association of impaired mineralization of bone and teeth and disturbed renal phosphate reabsorption with altered expression of Pex suggests that the Pex gene product may play a critical role in these processes.


Subject(s)
Bone and Bones/chemistry , Gene Expression Regulation/physiology , Hypophosphatemia, Familial/genetics , Proteins/genetics , RNA, Messenger/analysis , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA Mutational Analysis , DNA, Complementary/genetics , Endopeptidases/genetics , Female , Genetic Linkage , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Sequence Data , Organ Specificity , Osteoblasts/chemistry , PHEX Phosphate Regulating Neutral Endopeptidase , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , X Chromosome
7.
J Clin Endocrinol Metab ; 81(6): 2381-8, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8964881

ABSTRACT

Therapy for X-linked hypophosphatemia (XLH) only partially corrects skeletal lesions and is often complicated by hyperparathyroidism. 24,25(OH)2 D3 improves skeletal lesions in a murine model of XLH and suppresses PTH secretion in animals. Therefore, we undertook a placebo-controlled trial of 24,25(OH)2 D3 supplementation to standard treatment in patients with XLH to improve bone disease and reduce hyperparathyroid complications. Fifteen subjects with XLH receiving standard treatment [1,25(OH)2 D3 or dihydrotachysterol plus phosphate] were evaluated, supplemented with placebo, and reevaluated one yr later. 24,25(OH)2 D3 supplementation was then begun and studies repeated after another year. Each patient underwent a detailed evaluation of calcium homeostasis over a 24-h period. Rachitic abnormalities were assessed radiographically in children. Adults underwent bone biopsies. 24,25(OH)2 D3 normalized PTH values in nine subjects (peak PTH was 46.5 +/- 6.6 pmol/L at entry, 42.3 +/- 5.9 pmol/L after placebo, and 23.3 +/- 5.4 pmol/L after 24,25(OH)2 D3). Nephrogenous cAMP decreased at night, coincident with the decrease in PTH, and serum phosphorus was slightly greater with 24,25(OH)2 D3. Radiographic features of rickets improved during 24,25(OH)2 D3 supplementation in children, and osteoid surface decreased in adults. 24,25(OH)2 D3 is a useful adjunct to standard therapy in XLH by effecting correction of hyperparathyroidism and improvement of rickets and osteomalacia.


Subject(s)
24,25-Dihydroxyvitamin D 3/therapeutic use , Bone and Bones/drug effects , Genetic Linkage , Hyperparathyroidism/drug therapy , Hypophosphatemia, Familial/drug therapy , Hypophosphatemia, Familial/genetics , X Chromosome , 24,25-Dihydroxyvitamin D 3/adverse effects , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Prospective Studies , Single-Blind Method
8.
J Bone Miner Res ; 10(3): 424-31, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7785464

ABSTRACT

Bone cells isolated from the Hyp mouse, the murine homologue for hypophosphatemic vitamin D-resistant rickets, produce abnormal bone when transplanted to either normal or phosphate-supplemented Hyp mice. To assess whether correction of the bone formation by mutant cells transplanted into either normal or Hyp mice could be achieved in the presence of supraphysiologic serum concentrations of 1.25-dihydroxyvitamin D3 (1.25-(OH)2D3), recipient mice of both genotypes were infused continuously with 1.25-(OH)2D3 (0.2 micrograms/kg/day). Bone nodules present in transplants recovered after 14 days were characterized by measuring the osteoid thickness and volume. Administration of 1.25-(OH)2D3 to Hyp mice corrected the defective bone formation by normal cells but not by pair-transplanted Hyp cells, despite normalization of serum phosphate levels and 3-fold increases in serum 1.25-(OH)2D3. The osteoid thickness and volume in Hyp transplants into 1.25-(OH)2D3-treated Hyp mice were, however, markedly reduced down to values observed for Hyp transplants into recipient normal mice. Administration of 1.25-(OH)2D3 to normal mice improved further bone formation by mutant cells without affecting that by pair-transplanted normal cells. Administration of 24.25-(OH)2D3 (1 microgram/kg/day) combined with 1.25-(OH)2D3 to recipient mice of both genotypes prevented the sharp fall in serum 24.25-(OH)2D3 but was not more beneficial than 1.25-(OH)2D3 alone for improving bone formation by transplanted Hyp cells. These observations demonstrate an abnormal response of the mutant cells to the extracellular environment and support the concept of an intrinsic osteoblast defect in the Hyp mouse.


Subject(s)
Bone Development/drug effects , Calcitriol/pharmacology , Hypophosphatemia, Familial/drug therapy , 24,25-Dihydroxyvitamin D 3/blood , 24,25-Dihydroxyvitamin D 3/pharmacology , 24,25-Dihydroxyvitamin D 3/therapeutic use , Alkaline Phosphatase/blood , Analysis of Variance , Animals , Calcitriol/blood , Calcitriol/therapeutic use , Calcium/blood , Cell Transplantation , Drug Therapy, Combination , Female , Hypophosphatemia, Familial/genetics , Hypophosphatemia, Familial/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Phosphates/blood , Skull/cytology , Stem Cells/cytology , Stem Cells/drug effects
9.
Acta Paediatr Suppl ; 405: 117-22, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7734783

ABSTRACT

Fat and mineral metabolic balance studies were performed in 25 normal very low-birth-weight infants (< or = 1500 g at birth) fed either pooled pasteurized human milk supplemented with calcium, phosphorus and magnesium, or a preterm formula. Calcium, phosphorus and magnesium intake were similar in both groups and averaged 100 mg/kg/day, 72 mg/kg/day and 8 mg/kg/day, respectively. Calcium and phosphorus retention was higher in the subjects fed fortified human milk than in those receiving a preterm formula (65 +/- 14 and 62 +/- 9 mg/kg/day versus 55 +/- 12 and 47 +/- 7 mg/kg/day respectively). The difference was only significant for phosphorus. Magnesium retention was similar in the two groups and averaged 3 mg/kg/day. Fat intake and absorption was significantly higher in the preterm formula fed group than in the one fed fortified human milk (5.5 +/- 0.4 g/kg/day and 88 +/- 4% versus 4.2 +/- 1 g/kg/day, 79 +/- 6% respectively). Assessment of the whole body bone mineral content by dual energy X-ray absorptiometry was performed at 3 and 6 months of age in another group of 25 low-birth-weight infants fed either fortified human milk or a preterm formula. Whole body bone mineral content (BMCt) was low (43.3 +/- 30.8 g of hydroxyapatite) at 3 months of age (theoretical term) compared to normal full-term newborns at birth. There was no significant influence of the diet. At 6 months of age, BMCt reached 168.6 +/- 36.6 g, a value similar to that of full-term newborns, with no significant difference between the two regimen groups.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Bone Density , Infant, Low Birth Weight/metabolism , Minerals/metabolism , Calcium/metabolism , Food, Fortified , Humans , Infant , Infant, Newborn , Phosphorus/metabolism , Potassium/metabolism
10.
Bone ; 15(3): 321-8, 1994.
Article in English | MEDLINE | ID: mdl-7520724

ABSTRACT

Osteogenesis imperfecta (OI) is a group of inherited disorders characterized by a predisposition to bone fracturing, and usually resulting from mutations in the genes encoding type I collagen. This report describes the molecular defects in a patient with type II OI and another with type III OI. These patients were demonstrated to possess point mutations resulting in glycine-->arginine substitutions within the triple helical domain of the alpha 1(I) or alpha 2(I) collagen polypeptide chain. The defect in the type II OI patient affected residue 211 of the alpha 1(I) triple helical domain, and constitutes the most amino-terminal lethal glycine-->arginine substitution described to date. The substitution in the type III OI patient affected residue 427 of the alpha 2(I) triple helical domain. Both defects were informative in that they identified the regions of the alpha 1(I) and alpha 2(I) collagen chains in which the phenotypes associated with glycine-->arginine substitutions undergo a transition between lethal and nonlethal forms, thereby allowing a more reliable prognosis of disease severity. The histological examination of bone from these patients revealed striking abnormalities in the quantity and organization of mineralized bone structures, compared with age-matched controls. Although the patients were differently classified, no major differences in the magnitude of bone architectural changes could be perceived, consistent with the presence of their defects near a common phenotypic transition. The results are compatible with there being a gradient in severity between OI types II and III, and that parameters external to the gene mutations might account for the survival differences in the 2 cases presented in this study.


Subject(s)
Bone and Bones/pathology , Osteogenesis Imperfecta/genetics , Base Sequence , Cell Line , Child , Collagen/genetics , DNA, Complementary , Female , Femur/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Ilium/pathology , Molecular Sequence Data , Osteogenesis Imperfecta/pathology , Point Mutation , RNA/genetics
11.
J Bone Miner Res ; 7(5): 523-30, 1992 May.
Article in English | MEDLINE | ID: mdl-1319665

ABSTRACT

The hypophosphatemic (Hyp) mouse is the murine homolog for human hypophosphatemic vitamin D-resistant rickets. We previously reported that bone cells isolated from normal and Hyp mice produced abnormal bone when transplanted intramuscularly into mutant mice. To assess the role of hypophosphatemia on bone formation in transplants, normal and Hyp mouse periostea were pair transplanted into control or phosphate (P)-supplemented Hyp mice and into control or P-deprived normal mice. The bone nodules formed in transplants after 2 weeks were characterized by measuring the thickness of the surrounding osteoid seams and the relative osteoid volume. P restriction in normal recipient mice impaired bone formation by transplanted normal cells and aggravated the defective bone formation by Hyp cells. The osteoid thickness and volume remained significantly higher in Hyp transplants than in normal cotransplants, however. P supplementation of Hyp recipient mice normalized bone formation by transplanted normal cells but not by Hyp cells. However, a marked decrease in osteoid thickness and volume was observed in Hyp transplants down to values observed in normal recipient mice. These results indicate that hypophosphatemia is not the only cause of abnormal bone formation in the Hyp mouse but that an osteoblast dysfunction contributes to the bone disease. These observations further support the concept that the osteoblast may be an important target for the Hyp mutation.


Subject(s)
Bone Development/genetics , Diet , Genetic Linkage/genetics , Phosphates/deficiency , X Chromosome , Animals , Bone Transplantation/pathology , Bone Transplantation/physiology , Bone and Bones/cytology , Disease Models, Animal , Hypophosphatemia, Familial/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Phosphates/blood , Phosphates/therapeutic use , Reference Values
12.
J Pediatr ; 116(5): 765-72, 1990 May.
Article in English | MEDLINE | ID: mdl-2109792

ABSTRACT

Because the monobasic potassium phosphate salt (monobasic) improves the solubility of calcium and phosphorus in amino acid plus dextrose solutions, compared with the current mixtures of monobasic plus dibasic salts (dibasic), we tested the bioavailability and clinical effects of monobasic in 16 parenterally fed low birth weight infants at standard (n = 8) and high levels (n = 8) of mineral intakes. A constant infusion of macronutrients and vitamin D was provided in a crossover design of two four-day periods. With standard intakes of calcium (35 mg/kg/day, 0.9 mmol/kg/day) and phosphorus (30 mg/kg/day, 1 mmol/kg/day), there was no difference between monobasic and dibasic regimens on balance data or plasma biochemical monitoring (calcium, phosphorus, pH, carbon dioxide pressure, base excess, 1,25-dihydroxyvitamin D, 25-hydroxyvitamin D). With the use of the monobasic regimen, the mineral intakes were doubled without precipitation in the infusate: calcium, 70 mg/kg/day (1.8 mmol/kg/day), and phosphorus, 55 mg/kg/day (1.7 mmol/kg/day). This led to increased apparent retention of both calcium (63 +/- 5 mg/kg/day, 1.58 +/- 0.12 mmol/kg/day) and phosphorus (52 +/- 4 mg/kg/day, 1.67 +/- 0.14 mmol/kg/day) compared with that for standard levels of mineral intake. The improvement of calcium-phosphorus balance was accompanied by more severe calciuria (9 +/- 2 mg/kg/day, 0.2 +/- 0.05 mmol/kg/day) and by metabolic compensation for an increased acid load. In addition to the possibility of exceeding the buffering capacity of the infant, this relative acidosis could also be evidence of improved bone mineralization.


Subject(s)
Calcium/pharmacokinetics , Infant, Low Birth Weight/metabolism , Parenteral Nutrition, Total , Phosphates/pharmacology , Phosphorus/pharmacokinetics , Potassium Compounds , Potassium/pharmacology , Amino Acids/administration & dosage , Calcium/administration & dosage , Calcium/urine , Gestational Age , Glucose/administration & dosage , Humans , Hydrogen-Ion Concentration , Infant Nutritional Physiological Phenomena , Infant, Low Birth Weight/urine , Infant, Newborn , Phosphorus/administration & dosage , Phosphorus/urine , Solubility
13.
Biol Neonate ; 52 Suppl 1: 119-30, 1987.
Article in English | MEDLINE | ID: mdl-3327527

ABSTRACT

Perinatal metabolism of vitamin D was studied in premature babies with the aim of: (1) reporting the relationship between the pregnant mother and her preterm infant and the metabolism of vitamin D during the first weeks of life, and (2) assessing the effect of vitamin D metabolites on phosphorus calcium and magnesium intestinal absorption. There was only a positive correlation between plasma cord calcium and 25-hydroxyvitamin D levels and the mother's plasma levels at birth. During the hypocalcemic episode observed during the first week of life, vitamin D activation did occur, but later on rickets or osteomalacia cannot be due to the low levels of vitamin D metabolites in the preterm receiving an adequate dose of vitamin D (1,000-1,200 IU of D2). Calcitriol, the major metabolite of vitamin D, is acting on the intestine and promotes calcium absorption even in very tiny prematures. The pathogenesis of hypomineralization in the preterm infant is due to the low intake of calcium or phosphorus and/or poor absorption of calcium in the case of vitamin D deficiency.


Subject(s)
Infant, Premature/metabolism , Vitamin D/metabolism , Calcium/pharmacokinetics , Female , Humans , Hypocalcemia/metabolism , Infant, Newborn , Maternal-Fetal Exchange , Phosphorus/pharmacokinetics , Pregnancy
14.
J Pediatr ; 109(2): 328-34, 1986 Aug.
Article in English | MEDLINE | ID: mdl-3488384

ABSTRACT

We assessed whether modification of vitamin D nutritional status during the last trimester of pregnancy affects maternal and neonatal calcium homeostasis. At the end of the first trimester, 40 pregnant women were randomly assigned to either of two groups, and blood taken to assess the basal values of Ca, Pi, Mg, iPTH, 25-OHD, and 1,25(OH)2D. From the sixth month on, group 1 (+D) received 1000 IU vitamin D3 daily; group 2 (-D) served as control. At the time of delivery, maternal serum 25-OHD was higher in the +D group (P less than 0.0005). Ca, Pi, iPTH, and 1,25(OH)2D were not affected. At term, venous cord 25-OHD levels were also higher in the +D group (P less than 0.0005), and 1,25(OH)2D levels slightly lower (P less than 0.05), but neither Ca, Pi, nor iPTH differed between the two groups. Serum CaT dropped significantly (P less than 0.002) at 4 days of age in the infants from both groups, although to a lesser extent in these from the +D group (P less than 0.05). Circulating iPTH increased in both groups. Serum 25-OHD remained low in the -D group, and dropped slightly in the +D group; 1,25(OH)2D remained stable during the first 4 days of life in the -D group, and increased in the +D group (P less than 0.001). Our data demonstrate the importance of providing adequate maternal vitamin D stores to ensure better perinatal handling of calcium. This is of particular importance for populations at risk for hypovitaminosis D.


Subject(s)
Calcium/blood , Infant, Newborn , Pregnancy , Vitamin D/therapeutic use , 25-Hydroxyvitamin D 2 , Ergocalciferols/analogs & derivatives , Ergocalciferols/blood , Female , Fetal Blood/analysis , Homeostasis , Humans , Magnesium/blood , Parathyroid Hormone/blood , Phosphates/blood , Random Allocation
15.
Acta Paediatr Scand ; 73(3): 315-24, 1984 May.
Article in English | MEDLINE | ID: mdl-6547559

ABSTRACT

Eleven uremic children with osteodystrophy aged 3 to 17 years were studied during administration of 1,25-(OH)2D3 for periods up to 21 months. Nine children presented with pure hyperparathyroidism, one with osteomalacia and one with mixed bone disease. Bone biopsies were performed before initiation of therapy and after 6 to 21 months of treatment following double tetracycline labeling. Skeletal lesions were improved but not cured in 5 of 9 children with hyperparathyroidism. In three instances lesions remained unchanged and worsened in one. No significant change was observed in the child with osteomalacia. Moderate improvement was noted in the patient with mixed bone disease. The propensity to develop hypercalcemia was the major factor associated with treatment failure since it precluded administration of adequate amounts of medication. Therapy with 1,25-(OH)2D3 was associated with a spectacular improvement in growth velocity in two of six children under age twelve.


Subject(s)
Calcitriol/therapeutic use , Chronic Kidney Disease-Mineral and Bone Disorder/drug therapy , Adolescent , Alkaline Phosphatase/blood , Bone and Bones/metabolism , Bone and Bones/pathology , Child , Child, Preschool , Chronic Kidney Disease-Mineral and Bone Disorder/metabolism , Chronic Kidney Disease-Mineral and Bone Disorder/pathology , Female , Growth , Humans , Male , Parathyroid Hormone/blood , Phosphorus/blood
16.
J Cell Biol ; 96(3): 639-43, 1983 Mar.
Article in English | MEDLINE | ID: mdl-6833375

ABSTRACT

A method is presented for isolating osteoblasts from newborn mouse calvaria without the use of digestive enzymes. The procedure is based on the ability of osteoblasts to migrate from bone onto small glass fragments (Jones, S.J., and A. Boyde, 1977, Cell Tissue Res., 184:179-193). The isolated cells were cultured for up to 14 d in Dulbecco's modified Eagle's medium supplemented with 10% fetal calf serum and 50 micrograms/ml of ascorbic acid. 7-d cultures were incubated for 24 h with [3H]proline. High levels of collagen synthesis relative to total protein were found, as measured by collagenase digestion of medium and cell layer proteins. Analysis of pepsin-digested proteins from the same cultures by SDS PAGE showed that type I collagen was predominantly produced with small amounts of type III and V (alpha 1 chains) collagens. Osteoblasts grown in the presence of beta-glycerophosphate were able to initiate mineral deposition in culture. Electron microscopic analysis of the cultures revealed the presence of needle-shaped apatite-like crystals associated with collagen fibrils and vesicles in the extracellular space. Mouse skin fibroblasts cultured under identical conditions failed to initiate mineralization. Electron histochemical studies revealed the presence of alkaline phosphatase activity, associated with osteoblast membranes, matrix vesicles and on or near collagen fibrils. Thus these isolated osteoblasts retained in culture their unique property of initiating mineralization and therefore represent a model of value for studying the mineralization process in vitro.


Subject(s)
Bone Matrix/physiology , Cells, Cultured/physiology , Osteoblasts/physiology , Osteogenesis , Animals , Cell Separation , Collagen/biosynthesis , Fibroblasts/physiology , Glycerophosphates/pharmacology , Mice , Mice, Inbred C57BL , Skull
17.
Acta Paediatr Scand ; 72(2): 203-6, 1983 Mar.
Article in English | MEDLINE | ID: mdl-6601357

ABSTRACT

In order to evaluate after birth the changes in circulating vitamin D metabolite levels in preterm babies supplemented with vitamin D (2 100 I.U./d), the serum concentration of 25-hydroxyvitamin D [25-OHD] and 1 alpha,25-dihydroxyvitamin D [1,25(OH)2D] were measured in 22 infants (31 to 35 weeks of gestation) from birth up to 96 hours of age. Compared to cord blood levels, serum calcium decreased significantly during the first 24 hours of life (p less than 0.005) and remained low until day 4. Serum immunoreactive parathyroid hormone (iPTH) levels increased from birth to 24 hours and then plateaued. The 25-OHD levels at birth were 27.5 +/- 2.5 nmol/l and increased to 67.5 +/- 12.5 nmol/l (p less than 0.005) during the four days of the study. During the same period, the 1,25(OH)2D serum levels increased steadily from 84 less than 7 to 343 less than 105 pmol/l (p less than 0.005). At all times, there was a positive correlation between 25-OHD levels and those of 1,25(OH)2D. Our data demonstrate that in preterm infants after 31 weeks of gestation, absorption and activation of vitamin D is present as soon as 24 hours after birth and that early neonatal hypocalcemia is unlikely to be caused by an impairment of either PTH secretion or vitamin D activation.


Subject(s)
Dihydroxycholecalciferols/blood , Ergocalciferols/analogs & derivatives , Infant, Premature , Vitamin D/blood , 25-Hydroxyvitamin D 2 , Ergocalciferols/blood , Humans , Infant, Newborn , Vitamin D/administration & dosage
18.
Dev Pharmacol Ther ; 6(3): 157-61, 1983.
Article in English | MEDLINE | ID: mdl-6861601

ABSTRACT

Serum 25-hydroxyvitamin D (25-OHD) levels were measured in 74 children randomly treated in a double-blind fashion with either 5 phenobarbital/kg/day or placebo after a single febrile seizure. Phenobarbital treatment for a period from 5 to 12 months had no effect on the 25-OHD levels. However, both in the phenobarbital and the placebo groups, vitamin D supplementation raised the circulating 25-OHD concentration (p less than 0.05). Whether it has a protective effect remains thus to be seen.


Subject(s)
Calcifediol/blood , Phenobarbital/pharmacology , Child, Preschool , Humans , Infant
19.
N Engl J Med ; 307(10): 584-8, 1982 Sep 02.
Article in English | MEDLINE | ID: mdl-7110204

ABSTRACT

We performed a histomorphometric study of trabecular-bone formation and resorption in undecalcified sections of iliac crest from three children presenting with clinical, radiologic, and biochemical evidence of rickets associated with dietary calcium deficiency. All three children had severe osteomalacia documented by hyperosteoidosis and reduced static and dynamic indicators of bone mineralization. There was a reduction of the calcified bone volume associated with a decreased bone formation rate and features of increased bone resorption. Correction of dietary calcium intake in two of the patients led to normal serum and urinary calcium levels and reduced alkaline phosphatase levels. After calcium therapy, the calcified bone volume was normal and indicators of bone mineralization returned to normal. We conclude that low calcium intake in children may be associated with a histologic picture of severe osteomalacia. Our finding that adequate amounts of calcium rapidly improved bone mineralization demonstrates that calcium deficiency can cause osteomalacia in children.


Subject(s)
Calcium/deficiency , Osteomalacia/etiology , Adolescent , Alkaline Phosphatase/blood , Bone Resorption , Bone and Bones/pathology , Calcium/metabolism , Calcium, Dietary/administration & dosage , Child , Child, Preschool , Female , Humans , Magnesium/blood , Male , Osteomalacia/pathology , Phosphorus/blood , Rickets/etiology
20.
Endocrinology ; 111(3): 904-11, 1982 Sep.
Article in English | MEDLINE | ID: mdl-6896684

ABSTRACT

The X-linked hypophosphatemic (Hyp) mouse presents with biochemical and skeletal abnormalities similar to those of human vitamin D-resistant rickets and hence is considered as a model of the human disease. In an attempt to correct osteomalacia, young (21-day-old) mutant male mice were infused continuously for 4 weeks with 1,25-dihydroxyvitamin D3 [1,25-(OH)2D3; 0.05--0.25 microgram/kg . day]. Mineral and skeletal changes were assessed by serum, urinary, and bone ash concentrations of calcium, phosphorus, and magnesium and by histomorphometric analysis of bone formation measured on histological sections of tetracycline dual labeled undecalcified caudal vertebrae. Treatment with 1,25-(OH)2D3 produced a dose-dependent elevation of serum phosphorous that could be assigned to increased intestinal phosphate absorption. Concomitantly, epiphyseal, endosteal, and periosteal bone mineralization were improved in correlation with both the dosage of 1,25-(OH)2D3 and the serum phosphorus level. Normalization of serum calcium and phosphorus but not of urinary phosphate excretion were achieved together with complete healing of bone mineralization when the highest doses of 1,25-(OH)2D3 (0.175--0.35 microgram/kg . day) were given. The data show that rickets and osteomalacia, which characterize the young Hyp mouse, can be healed by 1,25-(OH)2D3 in doses high enough to normalize serum mineral concentrations. Unlike the renal phosphate leak, the phenotypic expression of the Hyp gene pertaining to bone mineralization is then corrected by 1,25-(OH)2D3 supplementation.


Subject(s)
Calcitriol/therapeutic use , Hypophosphatemia, Familial/complications , Osteomalacia/drug therapy , Animals , Calcium/blood , Magnesium/blood , Male , Mice , Osteomalacia/complications , Phosphates/therapeutic use , Phosphorus/blood , Weaning
SELECTION OF CITATIONS
SEARCH DETAIL