Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 138
Filter
1.
Medicine (Baltimore) ; 97(40): e12507, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30290606

ABSTRACT

INTRODUCTION: Phosphaturic mesenchymal tumor mixed connective tissue type (PMT/MCT) is the most common type (up to 90%) of phosphaturic mesenchymal tumor (PMT), a rare clinicopathologic entity. Besides overproduction of fibroblast growth factor 23 (FGF23), there is a big variation of immunohistochemical characteristic across types of PMT, which makes it difficult to obtain an early diagnosis of PMT/MCT. As a benign tumor, PMT/MCT usually happens in subcutaneous tissues and leads to nonhealing of wound. A complete excision of PMT/MCT facilitates wound healing. CONCLUSIONS: Review of the existing evidence indicates that early diagnosis of PMT/MCT is critically important when treating PMT/MCT wound. Hence standardization of early diagnosis for PMT/MCT is mandated.


Subject(s)
Hypophosphatemia, Familial/diagnosis , Mesenchymoma/diagnosis , Mixed Connective Tissue Disease/diagnosis , Soft Tissue Neoplasms/diagnosis , Wounds and Injuries/complications , Biomarkers, Tumor/blood , Diagnosis, Differential , Early Detection of Cancer , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/biosynthesis , Humans , Hypophosphatemia, Familial/blood , Hypophosphatemia, Familial/etiology , Mesenchymoma/blood , Mesenchymoma/etiology , Mixed Connective Tissue Disease/blood , Mixed Connective Tissue Disease/etiology , Soft Tissue Neoplasms/blood , Soft Tissue Neoplasms/etiology , Wounds and Injuries/blood
2.
Am J Physiol Renal Physiol ; 315(2): F353-F363, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29667913

ABSTRACT

Loss of ubiquitin COOH-terminal hydrolase L1 (UCHL1), a deubiquitinating enzyme required for neuronal function, led to hyperphosphatemia accompanied by phosphaturia in mice, while calcium homeostasis remained intact. We therefore investigated the mechanisms underlying the phosphate imbalance in Uchl1-/- mice. Interestingly, phosphaturia was not a result of lower renal brush border membrane sodium-phosphate cotransporter expression as sodium-phosphate cotransporter 2a and 2c expression levels was similar to wild-type levels. Plasma parathyroid hormone and fibroblast growth factor 23 levels were not different; however, fibroblast growth factor 23 mRNA levels were significantly increased in femur homogenates from Uchl1-/- mice. Full-length and soluble α-klotho levels were comparable in kidneys from wild-type and Uchl1-/- mice; however, soluble α-klotho was reduced in Uchl1-/- mice urine. Consistent with unchanged components of 1,25(OH)2D3 metabolism (i.e., CYP27B1 and CYP24A1), sodium-phosphate cotransporter 2b protein levels were not different in ileum brush borders from Uchl1-/- mice, suggesting that the intestine is not the source of hyperphosphatemia. Nonetheless, when Uchl1-/- mice were fed a low-phosphate diet, plasma phosphate, urinary phosphate, and fractional excretion of phosphate were significantly attenuated and comparable to levels of low-phosphate diet-fed wild-type mice. Our findings demonstrate that Uchl1-deleted mice exhibit perturbed phosphate homeostasis, likely consequent to decreased urinary soluble α-klotho, which can be rescued with a low-phosphate diet. Uchl1-/- mice may provide a useful mouse model to study mild perturbations in phosphate homeostasis.


Subject(s)
Diet , Glucuronidase/deficiency , Hyperphosphatemia/enzymology , Hypophosphatemia, Familial/enzymology , Kidney/enzymology , Phosphates/metabolism , Ubiquitin Thiolesterase/deficiency , Animals , Calcitriol/blood , Disease Models, Animal , Femur/metabolism , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Deletion , Genetic Predisposition to Disease , Glucuronidase/urine , Homeostasis , Hyperphosphatemia/blood , Hyperphosphatemia/genetics , Hyperphosphatemia/urine , Hypophosphatemia, Familial/blood , Hypophosphatemia, Familial/genetics , Hypophosphatemia, Familial/urine , Intestinal Absorption , Klotho Proteins , Mice, Knockout , Parathyroid Hormone/blood , Phenotype , Phosphates/blood , Phosphates/urine , Ubiquitin Thiolesterase/genetics
3.
J Clin Endocrinol Metab ; 99(2): 395-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24297794

ABSTRACT

CONTEXT: We report a case of tumor-induced osteomalacia with evidence of synchronous multifocal fibroblast growth factor 23 (FGF23) production. OBJECTIVE: The aim is to present a case of tumor-induced osteomalacia and to highlight the fact that incomplete removal of multifocal FGF23-producing tumors, which are not entirely picked up by functional imaging, could be the cause of treatment failure. SETTING: The patient was treated in the Department of Endocrinology of a tertiary care center in India. PATIENT: We report the case of a 42-year-old male with tumor-induced osteomalacia. INTERVENTION: We treated the tumor-induced osteomalacia with staged surgery of the two tumors. The 18F-fluorodeoxyglucose (FDG)-avid lesion (considered the sole culprit lesion after functional imaging) was resected first, followed by the non-FDG-avid lesion. The sequential removal of both tumors resulted in complete cure. RESULTS: The patient had hypophosphatemia and hyperphosphaturia. C-Terminal FGF23 level was elevated. Positron emission tomography-computed tomography showed two lesions-an FDG-avid lesion in the right leg, and a non-avid lesion in the left thigh. After removal of the FDG-avid lesion, the hypophosphatemia persisted, and the FGF23 level showed only modest reduction. The patient had complete clinical and biochemical resolution only after removal of the second non-FDG-avid tumor. CONCLUSIONS: We present the case of a tumor-induced osteomalacia whose biochemical parameters did not improve after removal of the FDG-avid tumor initially. The possibility of multifocal FGF23 production was considered, and the second, non-FDG-avid lesion was resected, which resulted in complete cure. Thorough clinical examination and meticulous follow-up with documentation of the biochemical resolution are necessary for management of all patients with this rare disorder.


Subject(s)
Hypophosphatemia, Familial/diagnosis , Hypophosphatemia/diagnosis , Neoplasms, Connective Tissue/diagnosis , Adult , Fibroblast Growth Factor-23 , Humans , Hypophosphatemia/blood , Hypophosphatemia/surgery , Hypophosphatemia, Familial/blood , Hypophosphatemia, Familial/surgery , India , Male , Middle Aged , Neoplasms, Connective Tissue/blood , Neoplasms, Connective Tissue/surgery , Osteomalacia , Paraneoplastic Syndromes
4.
PLoS One ; 7(7): e39229, 2012.
Article in English | MEDLINE | ID: mdl-22859939

ABSTRACT

The vast majority of glomerular filtrated phosphate is reabsorbed in the proximal tubule. Posttransplant phosphaturia is common and aggravated by sirolimus immunosuppression. The cause of sirolimus induced phosphaturia however remains elusive. Male Wistar rats received sirolimus or vehicle for 2 or 7 days (1.5mg/kg). The urine phosphate/creatinine ratio was higher and serum phosphate was lower in sirolimus treated rats, fractional excretion of phosphate was elevated and renal tubular phosphate reabsorption was reduced suggesting a renal cause for hypophosphatemia. PTH was lower in sirolimus treated rats. FGF 23 levels were unchanged at day 2 but lower in sirolimus treated rats after 7 days. Brush border membrane vesicle phosphate uptake was not altered in sirolimus treated groups or by direct incubation with sirolimus. mRNA, protein abundance, and subcellular transporter distribution of NaPi-IIa, Pit-2 and NHE3 were not different between groups but NaPi-IIc mRNA expression was lower at day 7. Transcriptome analyses revealed candidate genes that could be involved in the phosphaturic response. Sirolimus caused a selective renal phosphate leakage, which was not mediated by NaPi-IIa or NaPi-IIc regulation or localization. We hypothesize that another mechanism such as a basolateral phosphate transporter may be responsible for the sirolimus induced phosphaturia.


Subject(s)
Hypophosphatemia, Familial/chemically induced , Immunosuppressive Agents/pharmacology , Kidney Tubules, Proximal/drug effects , Sirolimus/pharmacology , Sodium-Phosphate Cotransporter Proteins/metabolism , Animals , Creatinine/blood , Creatinine/urine , Gene Expression/drug effects , Homeostasis , Hypophosphatemia, Familial/blood , Hypophosphatemia, Familial/urine , Immunosuppressive Agents/adverse effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/physiopathology , Male , Phosphates/blood , Phosphates/urine , Protein Transport/drug effects , Rats , Rats, Wistar , Sirolimus/adverse effects , Sodium-Phosphate Cotransporter Proteins/genetics
5.
J Bone Miner Res ; 26(6): 1295-302, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21611969

ABSTRACT

Tumor-induced osteomalacia (TIO) is characterized by renal phosphate wasting, hypophosphatemia, and aberrant vitamin D(3) metabolism and is caused by fibroblast growth factor 23 (FGF-23)-producing mesenchymal tumors, which are often difficult to locate. We investigated the utility of selective venous sampling in tumor localization. The primary endpoint was identification of the FGF-23 concentration ratio between the venous drainage of the tumor bed and the general circulation that was diagnostic of the location of an FGF-23-secreting tumor. Fourteen subjects underwent 15 sampling procedures after functional and anatomic imaging studies. Subjects fit into three imaging categories: no suspicious site, multiple sites, and single site (positive controls). FGF-23 levels were measured by ELISA. Suspicious tumors were resected for diagnosis, confirmation, and cure. In subjects with a positive venous sampling study and subsequent cure, a minimum ratio of 1.6 was diagnostic. In 7 of 14 subjects there was suggestive imaging, a diagnostic ratio, and an associated TIO tumor (true positive). Four of these required complicated resection procedures. In 4 of 14 subjects with no suspicious site on imaging studies, an FGF-23 diagnostic ratio was not detected (true negative). Biopsy or resection of a single lesion in 2 of 14 subjects with a diagnostic ratio failed to identify a TIO tumor (false positive). A diagnostic FGF-23 ratio was absent in 1 of 14 subjects whose tumor was a single highly suspicious lesion on imaging studies (false negative). These data yield a sensitivity of 0.87 [95% confidence interval (CI) 0.47-0.99] and a specificity of 0.71 (95% CI 0.29-0.96). Selective venous sampling for FGF-23 was particularly useful in subjects with multiple suspicious sites or an anatomically challenging planned resection but not in the absence of a suspicious lesion on imaging studies.


Subject(s)
Catheterization , Hypophosphatemia, Familial/blood , Hypophosphatemia, Familial/complications , Mesoderm/pathology , Neoplasms, Connective and Soft Tissue/blood , Neoplasms, Connective and Soft Tissue/diagnosis , Adolescent , Adult , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/blood , Humans , Male , Middle Aged , Neoplasms, Connective and Soft Tissue/complications , Neoplasms, Connective and Soft Tissue/diagnostic imaging , Positron-Emission Tomography , Treatment Outcome , Veins
6.
J Obstet Gynaecol Res ; 37(10): 1470-3, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21564408

ABSTRACT

Hypophosphatasia is an inheritable disorder characterized by defective bone mineralization and a deficiency of tissue-nonspecific alkaline phosphatase (TNSALP) activity. Screening for mutations in the TNSALP gene allows genetic counseling and prenatal diagnosis of the disease in families with severe forms of hypophosphatasia. A 33-year-old, gravida 4, para 3 Japanese woman was referred to Nagoya City University Hospital for prenatal genetic counseling because of two previous occurrences of fetal bone anomalies. The molecular examination showed that the fetus was homozygous for the TNSALP gene mutation c.1559delT, each parent being heterozygous. Genetic counseling was offered and at the next pregnancy, chorionic villus sampling was performed, whereupon genetic analysis confirmed that the fetus did not carry the familial mutation c.1559delT. Postnatal molecular genetic analysis using the cord tissue can provide a diagnosis of lethal hypophosphatasia and prenatal genetic diagnosis of the TNSALP gene allows time for parental counseling and delivery planning.


Subject(s)
Alkaline Phosphatase/genetics , Hypophosphatemia, Familial/diagnosis , Alkaline Phosphatase/blood , Calcification, Physiologic/genetics , Chorionic Villi Sampling , Female , Genetic Testing , Humans , Hypophosphatemia, Familial/blood , Hypophosphatemia, Familial/genetics , Pregnancy , Prenatal Diagnosis
7.
Calcif Tissue Int ; 84(2): 112-7, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19148564

ABSTRACT

Fibroblast growth factor 23 (FGF23), a recently discovered phosphaturic substance playing a key role in genetic and oncogenic phosphate diabetes, is involved in the physiological regulation of phosphate metabolism. Moderate idiopathic phosphate diabetes (IPD) leading to male osteoporosis and diffuse pain resembling fibromyalgia has been described. The aim of our study was to define the role of FGF23 in the mechanism of IPD. The study concerned 29 patients with IPD, mean age 53 +/- 11 years, of whom 72% were men. Fifteen subjects without bone disease and with normal serum phosphate and calcium levels were used as controls. Phosphate diabetes was confirmed by phosphate reabsorption level <85% and phosphate reabsorption threshold (TmPO4/GFR) <0.83. Known causes of phosphate diabetes were excluded. Fasting level of FGF23, serum phosphate, 1-25(OH)2D3, and parathyroid hormone were measured in patients and compared with FGF23 and serum phosphate in healthy controls. Spinal and hip bone mineral density (BMD) were measured by osteodensitometry. Sixteen of 29 patients had diffuse pain, 10 had osteoporosis according to the World Health Organization criteria, and 11 had osteopenia. Serum phosphate was significantly lower in patients than in controls, but FGF23 levels did not differ. Compared to patients with normal bone status, patients with osteopenia and osteoporosis had significantly decreased FGF23 levels, whereas serum phosphate was identical in the two groups. In all patients, serum phosphate and FGF23 were positively correlated and FGF23 and 1-25(OH)2D3 were negatively correlated. FGF23 seems not be a cause of IPD, and the FGF23/phosphate/1-25(OH)2D3 axis appeared to be functional.


Subject(s)
Fibroblast Growth Factors/blood , Hypophosphatemia, Familial/blood , Adult , Calcium/blood , Calcium/metabolism , Cross-Sectional Studies , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Humans , Hypophosphatemia, Familial/metabolism , Male , Middle Aged , Parathyroid Hormone/blood , Parathyroid Hormone/metabolism , Phosphates/blood , Phosphates/metabolism , Prospective Studies
9.
Am J Physiol Endocrinol Metab ; 291(1): E38-49, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16449303

ABSTRACT

Inactivating mutations of the PHEX (phosphate-regulating gene with homologies to endopeptidases on the X chromosome) endopeptidase, the disease-causing gene in X-linked hypophosphatemia (XLH), results in increased circulating levels of fibroblastic growth factor-23 (FGF23), a bone-derived phosphaturic factor. To determine the causal role of FGF23 in XLH, we generated a combined Fgf23-deficient enhanced green fluorescent protein (eGFP) reporter and Phex-deficient Hyp mouse model (Fgf23(+/-)/Hyp). eGFP expression was expressed in osteocytes embedded in bone that exhibited marked upregulation of eGFP in response to Phex deficiency and in CD31-positive cells in bone marrow venules that expressed low eGFP levels independently of Phex. In bone marrow stromal cells (BMSCs) derived from Fgf23(-/-)/Hyp mice, eGFP expression was also selectively increased in osteocyte-like cells within mineralization nodules and detected in low levels in CD31-positive cells. Surprisingly, eGFP expression was not increased in cell surface osteoblasts, indicating that Phex deficiency is necessary but not sufficient for increased Fgf23 expression in the osteoblast lineage. Additional factors, associated with either osteocyte differentiation and/or extracellular matrix, are necessary for Phex deficiency to stimulate Fgf23 gene transcription in bone. Regardless, the deletion of Fgf23 from Hyp mice reversed the hypophosphatemia, abnormal 1,25(OH)(2)D(3) levels, rickets, and osteomalacia associated with Phex deficiency. These results suggest that Fgf23 acts downstream of Phex to cause both the renal and bone phenotypes in Hyp mice.


Subject(s)
Fibroblast Growth Factors/metabolism , Hypophosphatemia, Familial/metabolism , Membrane Glycoproteins/metabolism , Metalloendopeptidases/metabolism , Osteocytes/physiology , Animals , Body Weight/physiology , Bone Density/physiology , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Calcium/blood , Cholecalciferol/blood , Disease Models, Animal , Female , Femur/metabolism , Femur/physiology , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/genetics , Gene Expression Regulation , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Hypophosphatemia, Familial/blood , Hypophosphatemia, Familial/genetics , Male , Membrane Glycoproteins/genetics , Metalloendopeptidases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Osteocytes/metabolism , PHEX Phosphate Regulating Neutral Endopeptidase , Parathyroid Hormone/blood , Phosphorus/blood , Promoter Regions, Genetic
10.
Horm Res ; 65(2): 83-8, 2006.
Article in English | MEDLINE | ID: mdl-16424674

ABSTRACT

BACKGROUND/AIMS: Hypocalcemic vitamin D-resistant rickets (HVDRR) is a rare monogenic autosomal recessive disorder associated with mutations in the gene of the vitamin D receptor (VDR), the mediator of 1,25(OH)2D3 action. Although many investigations have discussed the clinical manifestations and molecular etiology of this disease, only a few have investigated the biochemical and hormonal status of heterozygous HVDRR. The aim of the current work was to investigate the profile of selected biochemical and hormonal parameters related to the vitamin D endocrine system in a large number of HVDRR heterozygotes. METHODS: 67 relatives of 2 HVDRR patients, all members of an extended Greek kindred of five generations with a common ancestor, were included in the study. Direct sequencing was used to identify VDR gene mutations. Serum Ca, P, 25(OH)D, iPTH, and 1,25(OH)2D levels were determined in all members of the kindred. RESULTS: DNA analysis of the participants led to the design of two study groups: the HVDRR carriers (24) and the control subjects (43). Our results showed elevated circulating serum levels of 1,25(OH)2D3 and lower levels of PTH than their age- and sex-matched controls. No hypocalcemia or hypophosphatemia were detected in HVDRR carriers. CONCLUSIONS: Our findings suggest that HVDRR carriers may have compensatory elevated serum levels of 1,25(OH)2D3 through which they restrain PTH secretion. The study of HVDRR carriers could be a useful tool for the investigation of the vitamin D endocrine system.


Subject(s)
Calcium/blood , Heterozygote , Hypophosphatemia, Familial/blood , Vitamin D/analogs & derivatives , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Genetic Carrier Screening , Greece/epidemiology , Humans , Hypophosphatemia, Familial/genetics , Male , Middle Aged , Pedigree , Vitamin D/blood
11.
Endocrinology ; 146(12): 5358-64, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16123154

ABSTRACT

Fibroblast growth factor-23 (FGF-23) is a novel circulating peptide that regulates phosphorus (Pi) and vitamin D metabolism, but the mechanisms by which circulating FGF-23 itself is regulated are unknown. To determine whether the serum FGF-23 concentration is regulated by dietary intake of Pi, we fed wild-type (WT), Npt2a gene-ablated (Npt2a(-/-)), and Hyp mice diets containing varying Pi contents (0.02-1.65%). In WT mice, increases in dietary Pi intake from 0.02-1.65% induced a 7-fold increase in serum FGF-23 and a 3-fold increase in serum Pi concentrations. Across the range of dietary Pi, serum FGF-23 concentrations varied directly with serum Pi concentrations (r(2) = 0.72; P < 0.001). In Npt2a(-/-) mice, serum FGF-23 concentrations were significantly lower than in WT mice, and these differences could be accounted for by the lower serum Pi levels in Npt2a(-/-) mice. The serum concentrations of FGF-23 in Hyp mice were 5- to 25-fold higher than values in WT mice, and the values varied with dietary Pi intake. Fgf-23 mRNA abundance in calvaria was significantly higher in Hyp mice than in WT mice on the 1% Pi diet; in both groups of mice, fgf-23 mRNA abundance in calvarial bone was suppressed by 85% on the low (0.02%) Pi diet. In WT mice fed the low (0.02%) Pi diet, renal mitochondrial 1alpha-hydroxylase activity and renal 1alpha-hydroxylase (P450c1alpha) mRNA abundance were significantly higher than in mice fed the higher Pi diets and varied inversely with serum FGF-23 concentrations (r(2) = 0.86 and r(2) = 0.64; P < 0.001, respectively). The present data demonstrate that dietary Pi regulates the serum FGF-23 concentration in mice, and such regulation is independent of phex function. The data suggest that genotype-dependent and dietary Pi-induced changes in the serum FGF-23 concentration reflect changes in fgf-23 gene expression in bone.


Subject(s)
Fibroblast Growth Factors/metabolism , Phosphorus, Dietary/pharmacology , Phosphorus/blood , Vitamin D/analogs & derivatives , Animals , Bone and Bones/metabolism , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/genetics , Hypophosphatemia, Familial/blood , Kidney/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Osmolar Concentration , RNA, Messenger/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIa/deficiency , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Vitamin D/metabolism , Vitamin D3 24-Hydroxylase
12.
J Endocrinol ; 183(3): R1-9, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15590969

ABSTRACT

MEPE (Matrix Extracellular PhosphoglycoprotEin) expression is markedly elevated in X-linked-hypophosphatemic-rickets (HYP) and tumor-induced osteomalacia (TIO). In normal individuals, circulating serum-levels of MEPE are tightly correlated with serum-phosphorus, parathyroid hormone (PTH) and bone mineral density (BMD). Also, MEPE derived, C-terminal ASARM-peptides are candidate minhibins and/or phosphatonins. Our aims were to determine: 1. whether MEPE-ASARM-peptide(s) are abnormally elevated in HYP/hyp serum, and, 2. whether the ASARM-peptide(s) accumulate in hyp mice kidney renal-tubules. Using a specific competitive ELISA we measured a five fold increase (P=0.007) of serum ASARM-peptide(s) in human HYP patients (normal subjects 3.25 microM n=9; S.E.M.=0.51 and HYP-patients 15.74 microM, n=9; S.E.M.=3.32). A 6.23 fold increase (P=0.008) was measured in hyp male mice compared with their normal male siblings (normal-siblings, 3.73 muM, S.E.M.=0.57, n=3; and hyp-mice 23.4 microM, n=3, S.E.M.=4.01). Renal immuno-histological screening also revealed a dramatic increase of ASARM-peptides in regions anatomically consistent with the proximal convoluted tubules. This study demonstrates for the first time that markedly elevated serum levels of protease-resistant ASARM-peptide(s) occur in HYP/hyp and they accumulate in murine hyp kidneys. These peptides are thus likely responsible for the phosphaturia and defective mineralization in HYP/hyp and TIO.


Subject(s)
Extracellular Matrix Proteins/blood , Glycoproteins/blood , Hypophosphatemia, Familial/blood , Phosphoproteins/blood , Animals , Case-Control Studies , Enzyme-Linked Immunosorbent Assay/methods , Extracellular Matrix Proteins/analysis , Female , Glycoproteins/analysis , Humans , Immunohistochemistry/methods , Kidney Tubules/chemistry , Male , Mice , Phosphates/urine , Phosphoproteins/analysis
14.
Endocrinology ; 145(11): 5269-79, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15284207

ABSTRACT

Fibroblast growth factor 23 (FGF23) is a recently characterized protein likely involved in the regulation of serum phosphate homeostasis. Increased circulating levels of FGF23 have been reported in patients with renal phosphate-wasting disorders, but it is unclear whether FGF23 is the direct mediator responsible for the decreased phosphate transport at the proximal renal tubules and the altered vitamin D metabolism associated with these states. To examine this question, we generated transgenic mice expressing and secreting from the liver human FGF23 (R176Q), a mutant form that fails to be degraded by furin proteases. At 1 and 2 months of age, mice carrying the transgene recapitulated the biochemical (decreased urinary phosphate reabsorption, hypophosphatemia, low serum 1,25-dihydroxyvitamin D(3)) and skeletal (rickets and osteomalacia) alterations associated with these disorders. Unexpectantly, marked changes in parameters of calcium homeostasis were also observed, consistent with secondary hyperparathyroidism. Moreover, in the kidney the anticipated alterations in the expression of hydroxylases associated with vitamin D metabolism were not observed despite the profound hypophosphatemia and increased circulating levels of PTH, both major physiological stimuli for 1,25-dihydroxyvitamin D(3) production. Our findings strongly support the novel concept that high circulating levels of FGF23 are associated with profound disturbances in the regulation of phosphate and vitamin D metabolism as well as calcium homeostasis and that elevated PTH levels likely also contribute to the renal phosphate wasting associated with these disorders.


Subject(s)
Fibroblast Growth Factors/genetics , Hypophosphatemia, Familial/physiopathology , Osteomalacia/physiopathology , Parathyroid Hormone/physiology , Animals , Bone and Bones/pathology , Bone and Bones/physiopathology , Calcium/blood , Carcinoma, Hepatocellular , Cell Line, Tumor , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/blood , Gene Expression , Humans , Hypophosphatemia, Familial/blood , Hypophosphatemia, Familial/pathology , Kidney Tubules, Proximal/physiopathology , Male , Mice , Mice, Transgenic , Osteomalacia/blood , Osteomalacia/pathology , Parathyroid Hormone/blood , Phosphates/blood , Pregnancy , Vitamin D/blood
16.
Bone ; 34(1): 134-9, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14751570

ABSTRACT

X-linked hypophosphatemia (XLH) is characterized by rickets and osteomalacia and arises from mutations in the Phex and PHEX genes in mice (Hyp) and humans, respectively. The present study was undertaken to examine the effect of gene dose on the skeletal phenotype using a histomorphometric approach. Metrical traits (vertebral length, growth plate thickness, cancellous osteoid volume per bone volume, and cancellous, endocortical, and periosteal osteoid thickness) were compared in caudal vertebrae of mutant female (Hyp/+, Hyp/Hyp) and male (Hyp/Y) mice and their normal female (+/+) and male (+/Y) littermates. Mutant animals had trait values that differed significantly from those of normal animals. However, with the exception of vertebral length and cancellous osteoid thickness, values were not significantly different between the three mutant genotypes. We also examined the effect of gamete-of-origin on histomorphometric parameters in obligate Hyp/+ females derived from male or female transmitting parents. The metrical trait values in both groups of Hyp/+ mice were similar, with the exception of vertebral length and cancellous osteoid volume per bone volume. In summary, we demonstrate that the amount of osteoid per bone volume is similar in the three mutant genotypes and conclude that the extent and magnitude of the mineralization defect is fully dominant and likely not affected by gene dose. The differences in vertebral length in the mutants suggest that rickets and osteomalacia are not the only causes of decreased vertebral growth in Hyp mice and that Phex protein may influence bone growth and mineralization by distinct pathways.


Subject(s)
Bone and Bones/pathology , Gene Dosage , Genetic Diseases, X-Linked/genetics , Hypophosphatemia, Familial/genetics , Hypophosphatemia, Familial/pathology , Proteins/genetics , Proteins/metabolism , Animals , Bone and Bones/metabolism , Breeding , Female , Genetic Diseases, X-Linked/metabolism , Genetic Diseases, X-Linked/pathology , Genotype , Hypophosphatemia, Familial/blood , Hypophosphatemia, Familial/metabolism , Male , Mice , Mice, Mutant Strains , Mice, Transgenic , Mutation/genetics , PHEX Phosphate Regulating Neutral Endopeptidase , Phosphates/blood , X Chromosome/genetics
19.
J Bone Miner Res ; 18(7): 1227-34, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12854832

ABSTRACT

UNLABELLED: We investigated if the circulating levels of the phosphaturic factor FGF23 are elevated in subjects with XLH. Although we failed to find a statistically significant increase, FGF23 levels were significantly correlated with the degree of hypophosphatemia in XLH. In contrast, FGF23 levels were markedly increased in subjects with ESRD and correlated inversely with the degree of hyperphosphatemia. INTRODUCTION: Inactivating mutations of PHEX cause renal phosphate wasting in X-linked hypophosphatemic rickets (XLH) because of the accumulation of a phosphaturic hormone called phosphatonin. The recent discovery that FGF23 is the circulating phosphaturic factor in autosomal dominant hypophosphatemia raises the possibility that FGF23 is phosphatonin. METHODS: Fasting serum FGF23 levels and serum biochemical parameters were measured using a human FGF23 (C-terminal) ELISA assay in 11 subjects with XLH and 42 age-matched controls, 5 subjects with hypophosphatemia of unknown cause, and 14 hyperphosphatemic subjects with end stage renal disease (ESRD). Associations between variables were examined using the Spearman's correlation coefficient and linear regression analysis. RESULTS AND CONCLUSIONS: FGF23 (RU/ml) concentrations were not different (p = 0.11) between control and hypophosphatemic XLH subjects, but were significantly increased in hyperphosphatemic subjects with ESRD (p < 0.001). Western blot analysis found the presence of both full-length and C-terminal FGF23 fragments in serum from ESRD subjects. There was a strong inverse correlation between FGF23 and serum phosphorus (r = -0.60) and calcium and phosphorus (Ca x P) product (r = -0.65) in XLH, and a strong positive relationship between FGF23 and Pi (r = 0.50) and Ca x P product (r = 0.62) in ESRD. FGF23 levels were variably elevated in subjects with hypophosphatemia of unknown cause, one of which had tumor-induced osteomalacia (TIO). Removal of the tumor resulted in rapid reduction in serum FGF23 levels. These findings suggest that FGF23 has a possible role in mediating hypophosphatemia in XLH and TIO, but the overlapping levels of FGF23 in hypophosphatemic disorders and normal subjects indicate that serum phosphorus and FGF23 can also be independently regulated.


Subject(s)
Fibroblast Growth Factors/blood , Homeostasis , Hypophosphatemia, Familial/blood , Hypophosphatemia/blood , Kidney Failure, Chronic/blood , Phosphorus/metabolism , Adult , Blotting, Western , Case-Control Studies , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Fasting/blood , Female , Fibroblast Growth Factor-23 , Humans , Male , Middle Aged , Phosphorus/blood
20.
N Engl J Med ; 348(17): 1656-63, 2003 Apr 24.
Article in English | MEDLINE | ID: mdl-12711740

ABSTRACT

BACKGROUND: Mutations in fibroblast growth factor 23 (FGF-23) cause autosomal dominant hypophosphatemic rickets. Clinical and laboratory findings in this disorder are similar to those in oncogenic osteomalacia, in which tumors abundantly express FGF-23 messenger RNA, and to those in X-linked hypophosphatemia, which is caused by inactivating mutations in a phosphate-regulating endopeptidase called PHEX. Recombinant FGF-23 induces phosphaturia and hypophosphatemia in vivo, suggesting that it has a role in phosphate regulation. To determine whether FGF-23 circulates in healthy persons and whether it is elevated in those with oncogenic osteomalacia or X-linked hypophosphatemia, an immunometric assay was developed to measure it. METHODS: Using affinity-purified, polyclonal antibodies against [Tyr223]FGF-23(206-222)amide and [Tyr224]FGF-23(225-244)amide, we developed a two-site enzyme-linked immunosorbent assay that detects equivalently recombinant human FGF-23, the mutant form in which glutamine is substituted for arginine at position 179 (R179Q), and synthetic human FGF-23(207-244)amide. Plasma or serum samples from 147 healthy adults (mean [+/-SD] age, 48.4+/-19.6 years) and 26 healthy children (mean age, 10.9+/-5.5 years) and from 17 patients with oncogenic osteomalacia (mean age, 43.0+/-13.3 years) and 21 patients with X-linked hypophosphatemia (mean age, 34.9+/-17.2 years) were studied. RESULTS: Mean FGF-23 concentrations in the healthy adults and children were 55+/-50 and 69+/-36 reference units (RU) per milliliter, respectively. Four patients with oncogenic osteomalacia had concentrations ranging from 426 to 7970 RU per milliliter, which normalized after tumor resection. FGF-23 concentrations were 481+/-528 RU per milliliter in those with suspected oncogenic osteomalacia and 353+/-510 RU per milliliter (range, 31 to 2335) in those with X-linked hypophosphatemia. CONCLUSIONS: FGF-23 is readily detectable in the plasma or serum of healthy persons and can be markedly elevated in those with oncogenic osteomalacia or X-linked hypophosphatemia, suggesting that this growth factor has a role in phosphate homeostasis. FGF-23 measurements might improve the management of phosphate-wasting disorders.


Subject(s)
Fibroblast Growth Factors/blood , Hypophosphatemia, Familial/blood , Osteomalacia/blood , Paraneoplastic Syndromes/blood , Adolescent , Adult , Amino Acid Sequence , Child , Child, Preschool , Enzyme-Linked Immunosorbent Assay/methods , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/genetics , Genetic Diseases, X-Linked/blood , Humans , Hypophosphatemia, Familial/genetics , Male , Middle Aged , Molecular Sequence Data , Mutation , Reference Values , Rickets/blood , Rickets/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...