当前位置: 首页 > 期刊 > 《美国生理学杂志》 > 2005年第6期 > 正文
编号:11416658
"Phosphatonins" and the regulation of phosphorus homeostasis
http://www.100md.com 《美国生理学杂志》
     1Division of Nephrology and Hypertension, Departments of Medicine and Biochemistry and Molecular Biology, Mayo Clinic Rochester, Mayo College of Medicine, Rochester, Minnesota

    2Receptor Ligand Therapeutics, Genzyme Corporation, Framingham, Massachusetts

    ABSTRACT

    Phosphate ions are critical for normal bone mineralization, and phosphate plays a vital role in a number of other biological processes such as signal transduction, nucleotide metabolism, and enzyme regulation. The study of rare disorders associated with renal phosphate wasting has resulted in the discovery of a number of proteins [fibroblast growth factor 23 (FGF-23), secreted frizzled related protein 4 (sFRP-4), matrix extracellular phosphoglycoprotein, and FGF 7 (FGF-7)] that decrease renal sodium-dependent phosphate transport in vivo and in vitro. The "phosphatonins," FGF-23 and sFRP-4, also inhibit the synthesis of 1,25-dihydroxyvitamin D, leading to decreased intestinal phosphate absorption and further reduction in phosphate retention by the organism. In this review, we discuss the biological properties of these proteins, alterations in their concentrations in various clinical disorders, and their possible physiological role.

    phosphate; secreted frizzled related protein 4; fibroblast growth factor 23; matrix extracellular phosphoglycoprotein; vitamin D

    PHOSPHATE HOMEOSTASIS IN MAMMALS

    Importance of Phosphorus in Biological Processes

    PHOSPHORUS PLAYS AN IMPORTANT role in a number of biological processes and is an exceptionally important component of hydroxyapatite, the major component of bone mineral (88). In addition, phosphorus is present in nucleic acids, bioactive signaling proteins, phosphorylated enzymes, and cell membranes (27, 51, 52, 63). A prolonged deficiency of phosphorus and inorganic phosphate results in serious biological problems, including impaired mineralization of bone resulting in osteomalacia or rickets, abnormal erythrocyte, leukocyte, and platelet function; impaired cell membrane integrity that can result in rhabdomyolysis; and impaired cardiac output (61, 62, 66, 67, 101). Therefore, the maintenance of appropriate phosphorus homeostasis is critical for the well-being of the organism.

    During skeletal growth and bone remodeling, calcium and phosphate are required for the formation of hydroxyapatite and other mineral-phase components. The rate at which mineralization occurs is dependent, in part, on the availability of phosphorus and calcium. In the absence of these ions, mineralization is impaired, resulting in the formation of poorly mineralized bone that is characteristic of osteomalacia or rickets (68). This defect is clearly demonstrated by reduced tetracycline uptake and minimal separation of sequential tetracycline labels (Fig. 1). The amount of nonmineralized matrix is significantly increased in osteomalacia as a consequence of reduced mineralization (68).

    Phosphorus Metabolism in Humans

    The normal adult body contains 15–20 mol of phosphate, and males have slightly more phosphate than females (39). A majority of phosphate (80–90%) is present in bone, with the remainder distributed between other tissues and extracellular fluid. Phosphate is present as inorganic phosphate and in a variety of organic substances including sugars, phosphoproteins, phospholipids, and nucleic acids. All tissues absorb and secrete phosphate into the extracellular fluid, and phosphate moves between the extracellular fluid pool and bone. It is not surprising that multiple factors and regulatory mechanisms play a role in maintaining serum phosphate concentrations within a physiological range. Currently, our understanding of phosphate regulation is limited to the major pathways associated with intestine, bone, and kidney (Fig. 2).

    A normal diet adequate in phosphorus normally contains 20 mg·kg–1·day–1 (1,500 mg) of phosphorus. Of this, 16 mg·kg–1·day–1 (1,100 mg) are absorbed in the proximal intestine, predominantly in the jejunum. Approximately 3 mg·kg–1·day–1 (200 mg) are secreted into the intestine via pancreatic and intestinal secretions, giving a net phosphorus absorption of 13 mg·kg–1·day–1 (900 mg). Approximately 7 mg·kg–1·day–1 of phosphorus appear in the feces. Absorbed phosphorus enters the extracellular fluid pool and moves in and out of bone as needed (3 mg·kg–1·day–1, 200 mg). The rate of bone remodeling (both resorption and mineralization) is important in determining the concentration of serum phosphorus in the extracellular fluid pool. Plasma phosphorus is filtered in the glomerulus and enters the tubular fluid. Unlike plasma calcium, which is bound to plasma proteins and only partly filtered, plasma phosphorus is filtered almost completely at the glomerulus and enters the tubular fluid in approximately the same concentrations as are present in plasma (39). Approximately 13 mg·kg–1·day–1 (900 mg) of phosphorus, an amount equivalent to that absorbed in the intestine, are excreted in the urine. In states of phosphate equilibrium or balance and normal renal function, the amount of phosphorus appearing in urine is remarkably similar to that absorbed in the intestine and can serve as a rough approximation of the amount absorbed in the intestine. In states of phosphate deprivation resulting from inadequate intake of phosphorus or low absorption of phosphorus from the intestine, urinary phosphorus concentrations are low and serve as appropriate indicators of altered phosphorus regulation (39, 114, 115, 117).

    Factors Regulating Phosphorus Homeostasis

    There are a number of hormones involved in the control of phosphorus metabolism. Parathyroid hormone and 1,25(OH)2D3 are among the best understood. Concentrations of these hormones are regulated by phosphorus in a manner that is conducive to the maintenance of normal phosphate balance (4, 8, 17, 19–21, 25, 26, 30, 41, 50, 57, 58, 64, 65, 95, 97, 109–112, 116, 121–125). In addition, a number of other substances such as growth hormone and insulin-like growth factor 1, alter phosphorus balance although their circulating concentrations are not directly controlled by ambient phosphorus concentrations (65). Recent studies have identified several new factors that also play a role in the regulation of phosphorus transport and homeostasis (66, 67, 102, 119). These include the "phosphatonins" fibroblast growth factor 23 (FGF-23) and secreted frizzled related protein-4 (sFRP-4) that induce a state of negative phosphate balance directly, by inhibiting renal phosphate reabsorption in the proximal tubule, and indirectly, by inhibiting the synthesis of 1,25(OH)2D3 and reducing the intestinal absorption of phosphorus. Two recently described factors, fibroblast growth factor 7 (FGF-7) and matrix extracellular phosphoglycoprotein (MEPE), have been shown to inhibit phosphate transport in renal epithelial cells in culture, and in the case of matrix extracellular phosphorus glycoprotein, to induce phosphaturia in mice. FGF-7 and MEPE, however, have not been demonstrated to prevent compensatory increases in serum 1,25(OH)2D3 concentrations seen in hypophosphatemic states or to directly inhibit 25-hydroxyvitamin D -hydroxylase activity.

    Figure 3 summarizes the factors currently known that control phosphorus homeostasis in mammals. Parathyroid hormone, by virtue of its phosphaturic effect in the kidney (11, 26, 39, 81), decreases overall phosphate retention, whereas 1,25(OH)2D3 increases phosphate retention by enhancing the efficiency of phosphorus absorption in the intestine and in the kidney (116, 121–125). It should be noted that parathyroid hormone has two opposing effects. As noted above, parathyroid hormone increases urinary phosphate excretion. At the same time, it also increases the synthesis of 1,25(OH)2D3 by stimulating the activity of the 1-hydroxylase enzyme in the kidney (17, 30, 41). 1,25(OH)2D3, in turn, increases the efficiency of phosphorus absorption in the intestine and kidney. In contrast, the phosphatonins increase renal phosphate excretion and inhibit 25-hydroxyvitamin D 1-hydroxylase activity, thereby further decreasing the retention of phosphorus.

    In Figure 4 are shown physiological changes that are known to occur with low or high dietary intakes of phosphate. A decrease in serum phosphate concentrations, such as occurs following the ingestion of a diet low in phosphorus, results in increased ionized calcium concentrations, decreased parathyroid hormone secretion, and a subsequent decreased renal phosphate excretion, such that serum phosphate concentrations remain normal (11, 39, 81). At the same time, by parathyroid hormone-independent mechanisms, there is increased renal 25-hydroxyvitamin D 1-hydroxylase activity, increased 1,25(OH)2D3 synthesis, and increased phosphorus absorption in the intestine and reabsorption in the kidney (31, 43–45, 121–125). Conversely, with elevated phosphate intake, there are decreased calcium concentrations, increased parathyroid hormone release from the parathyroid gland, and increased renal phosphate excretion. Increased serum phosphate concentrations inhibit renal 25-hydroxyvitamin D 1-hydroxylase, and decrease 1,25(OH)2D3 synthesis. Reduced 1,25(OH)2D3 concentrations decrease intestinal phosphorus absorption as well as renal phosphate reabsorption. All of these factors tend to bring serum phosphate concentrations back into the normal range.

    Factors Influencing Intestinal Phosphate Absorption and Renal Tubular Phosphate Reabsorption

    The absorption of phosphate in the intestine is dependent on the amount and availability of phosphorus present in the diet (31, 43, 45, 121, 122). For example, a diet containing an exceptionally low amount of phosphorus results in increased phosphorus absorption in the proximal intestine, predominantly the jejunum. Phosphate binders, such as aluminum hydroxide, calcium salts, lanthanum, and sevelamar (Renagel), reduce the amount of available phosphorus, thereby decreasing phosphate absorption and serum phosphate levels. 1,25-(OH)2D increases the efficiency of phosphorus absorption in the jejunum, and, conversely, low 1,25-(OH)2D concentrations reduce the efficiency of phosphorus absorption in the intestine (122, 123).

    A variety of factors influence bulk phosphate reabsorption along the proximal convoluted and straight tubule of the kidney (11, 81). In animals with intact parathyroid glands, phosphate reabsorption along the proximal convoluted tubule exceeds fluid reabsorption, such that the phosphate concentration in the proximal tubules is 70% of the plasma level (11, 81). In the absence of parathyroid hormone, phosphate reabsorption increases and the phosphate concentration in the proximal tubule is 30% of that in plasma. There is little phosphate reabsorption in the proximal straight tubule in the presence of parathyroid hormone. However, in the absence of parathyroid hormone, phosphate is avidly reabsorbed along the proximal straight tubule, resulting in very low urinary excretion of phosphate (11, 81). Hormonal and nonhormonal factors that influence phosphate reabsorption along the proximal tubule include parathyroid hormone concentrations, sodium reabsorption, serum calcium concentrations, 1,25(OH)2D, serum bicarbonate concentrations, hypercapnia or hypocapnia, dopamine, and serotonin (11, 81). Table 1 summarizes the influence of some of these factors on the efficiency of phosphorus reabsorption by the proximal tubule.

    Sodium-Phosphate Cotransporters Mediate the Uptake of Phosphate in Intestinal and Renal Epithelial Cells

    The uptake of phosphate is mediated by sodium-phosphate cotransporters that are located at the apical border of intestinal cells (NaPi-IIb) and at apical borders of proximal tubule cells (NaPi-IIa and NaPi-IIc) (5, 12–15, 28, 48, 49, 59, 69, 76–80, 82–85, 90–92, 113, 129, 130, 132, 133). The structure and physiology of these cotransport molecules have been extensively reviewed, and the reader is directed to other publications in this regard (12, 14, 15, 76–84, 117). Suffice it to say that the sodium-phosphate cotransporters are highly homologous and are predicted to have similar structures (59, 83).

    Parathyroid hormone and 1,25(OH)2D maintain phosphate homeostasis through their regulation of the sodium-phosphate cotransporters in the kidney and intestine. The numbers of renal sodium-phosphate cotransporters is reduced along the apical borders of proximal tubular cells following the administration of parathyroid hormone 1–34, but not by the administration of parathyroid hormone 3–34, (129, 130). The renal sodium-phosphate cotransporter NaPi-IIa have been shown to be internalized and degraded within the lysosomes (49, 60, 78, 84).

    In the intestine, the number of Na/Pi-IIb cotransporters is increased within the apical membrane of absorptive cells following the administration of a diet low in phosphorus (48). Because phosphorus deprivation is also associated with an increase in the synthesis of 1,25(OH)2D, it is logical to assume that the increase in the number of transporters is due to the increased serum concentration of 1,25(OH)2D. The administration of 1,25(OH)2D is associated with an increase in the numbers of sodium-phosphate cotransporters present within apical membranes of intestinal cells (48). However, recent evidence suggests that in vitamin D receptor mutant mice, phosphate deprivation results in an upregulation in the numbers of transporters in intestinal cells even in the absence of vitamin D activity (103).

    "PHOSPHATONIN"

    Definition of Phosphatonin

    The term phosphatonin was introduced to describe a factor or factors responsible for the inhibition of renal phosphate reabsorption and altered 25-hydroxyvitamin D 1-hydroxylase regulation observed in patients with tumor-induced osteomalacia (35). Cai et al. (22) described a patient with tumor-induced osteomalacia in whom the biochemical characteristics of hypophosphatemia, renal phosphate wasting, and reduced 1,25(OH)2D, disappeared following removal of the tumor. Culture of tumor cells demonstrated the presence in tumor cell supernatants of 10- to 30-kDa heat-sensitive factor(s) that inhibited sodium-dependent phosphate transport (but not the transport of other substances such as glucose and amino acids) in opossum kidney cells, proximal tubular-like epithelial cells that have been previously shown to transport phosphate in a sodium-dependent manner. Unlike parathyroid hormone, this substance(s) did not increase intracellular cAMP concentrations. Furthermore, its activity was not blocked by parathyroid hormone receptor antagonists. When tumor cells were implanted in nude mice, the animals became hypophosphatemic within 3–6 mo following implantation of the cells. These observations suggested that a novel substance(s) capable of modulating phosphate transport in the proximal tubule cells was present in tumor cells. The term phosphatonin was coined to describe this circulating phosphaturic factor(s) that functions via cAMP-independent pathways and prevents or attenuates increased 25-hydroxyvitamin D 1-hydroxylase activity that normally occurs in response to hypophosphatemia.

    Biochemical Similarity Among Patients with Tumor-Induced Osteomalacia, X-Linked Hypophosphatemic Rickets, and Autosomal Dominant Hypophosphatemic Rickets

    A similar biochemical phenotype exists in patients with X-linked hypophosphatemic rickets (XLH) and the animal model, the Hyp mouse (32, 33, 36, 37, 93, 94). Several investigators have shown the presence of circulating factors in the serum of Hyp mice that inhibit sodium-dependent phosphate transport in the kidney (74, 75, 87). Hence, it was reasonable to assume that a factor similar to that found in patients with tumor-induced osteomalacia was also present in patients with XLH. Positional cloning experiments identified the mutant gene, but, surprisingly, the gene encoded an endopeptidase, PHEX, that resembled a protease responsible for the degradation of atrial natriuretic factor (2, 34, 73). It was postulated that the endopeptidase PHEX was responsible for the degradation of this phosphaturic peptide and that excessive amounts of this peptide circulated in serum when the endopeptidase was unable to degrade the peptide in question.

    Further interesting information became available when it was demonstrated that patients with the disease, autosomal dominant hypophosphatemic rickets, had activating mutations within the fibroblast growth factor homolog FGF-23 (1). The mutation in the FGF-23 gene resulted in the modification of a proconvertase site (176 RHTR 179) in the COOH-terminal region of the encoded peptide. Replacement of the arginine residues at either position 176 or 179 eliminated the cleavage site. Subsequent studies showed that the uncleaved mutant form of FGF-23 had a long half-life (7, 107, 134). Its persistence in the circulation resulted in the biochemical phenotype of hypophosphatemia, renal phosphate wasting, and low 1,25(OH)2D concentrations.

    PEPTIDE FACTORS RESPONSIBLE FOR TUMOR-INDUCED OSTEOMALACIA

    Identification of Phosphaturic Peptides from Tumors Responsible for Tumor-Induced Osteomalacia

    FGF-23 overexpression was also associated with tumor-induced osteomalacia (16, 106). FGF-23 infused or injected into mice was shown to induced hypophosphatemia, renal phosphate wasting, and reduced renal 25-hydroxyvitamin D 1-hydroxylase messenger RNA concentrations (101, 106). FGF-23 also inhibited sodium-dependent phosphate uptake in opossum kidney cells (16, 138). Furthermore, Bowe and co-workers (16) showed that FGF-23 was a substrate for PHEX inasmuch as recombinant PHEX degraded FGF-23 in vitro in rabbit reticulocyte lysate system. This latter observation has not been validated by others using somewhat different systems, although another report has shown that FGF-23-derived peptides are processed by PHEX (9, 23, 46).

    In addition to FGF-23, serial analysis of gene expression (SAGE) identified additional genes that were overexpressed in tumors associated with tumor-induced osteomalacia (29). Table 2 summarizes identified genes validated by other methods. Genes encoding secreted factors were expressed using recombinant methods and tested for phosphate-regulating activity in vitro. Similar to FGF-23, sFRP-4 was shown to inhibit sodium-dependent phosphate transport in opossum kidney cells and phosphate reabsorption in vivo in rats. In addition, infusion of sFRP-4 also prevented a compensatory increase in 25-hydroxyvitamin D 1-hydroxylase activity following the induction of hypophosphatemia. Thus sFRP-4 has the properties of a phosphatonin.

    Rowe and co-workers (98, 100) also isolated another molecule, MEPE, from tumors associated with tumor-induced osteomalacia (98, 100). This protein, when infused into mice, caused hypophosphatemia and renal phosphate wasting (100). Furthermore, it inhibits sodium-dependent phosphate transport in opossum kidney cells. Recently, Carpenter and co-workers (24) have also shown that FGF-7 is overexpressed in tumors associated with tumor-induced osteomalacia and may be a phosphaturic factor as it also inhibits phosphate transport in opossum kidney cells (24).

    In summary, using a variety of biochemical and molecular biological methods, at least four phosphate-regulating substances have been isolated from phosphate-wasting tumors associated with tumor-induced osteomalacia: FGF-23, sFRP-4, MEPE, and FGF-7. In the following sections, we will review the biological activity of the different phosphaturic peptides described above and discuss information concerning their role in various clinical disorders.

    FGF-23: BIOLOGICAL PROPERTIES AND ALTERATIONS IN CIRCULATING CONCENTRATIONS IN VARIOUS CLINICAL DISORDERS

    Biological Properties of FGF- 23

    As noted above, FGF-23 was initially postulated to be the factor responsible for autosomal dominant hypophosphatemic rickets (1). A mutation in the FGF-23 gene resulted in expression of an FGF-23 protein that was resistant to proteolysis and with a prolonged half-life and biopotency (7, 107, 134). Recombinant FGF-23 produced hypophosphatemia when administered intraperitoneally to mice (106). Serum calcium concentrations did not change following the administration of the peptide. When Chinese hamster ovary cells were transfected with an FGF-23 expression plasmid and the cells were implanted in nude mice, the animals became hypophosphatemic and the fractional excretion of phosphate was increased within 10 days. Alkaline phosphatase concentrations increased in the serum, consistent with changes in bone mineralization. Radiological evidence of rickets in the long bones and histological evidence of rachitic changes were observed after several weeks. There was a decrease in the amount of mRNA for the 25-hydroxyvitamin D 1-hydroxylase.

    In support of these studies, Bowe et al. (16) demonstrated that recombinant FGF-23 inhibited sodium-dependent phosphate transport in opossum kidney cells (see Fig. 5A). Furthermore, intravenous infusion of recombinant FGF-23 into mice caused a rapid, dose-dependent increase in the fractional excretion of phosphate with little or no change in sodium excretion (101) (Fig. 5B). These studies suggest that FGF-23 has direct actions on renal phosphate transport.

    The physiological role of FGF-23 in modulating plasma phosphate concentrations and 25 hydroxyvitamin D 1-hydroxylase levels was further supported by the generation of FGF-23 null mice (105). These mice had a reduced growth rate and died 10–14 wk after birth. Serum phosphate concentrations were elevated within 10 days after birth, and serum calcium concentrations became moderately elevated 2 wk after birth. Interestingly, these mice developed increased 25 hydroxyvitamin D 1-hydroxylase messenger RNA levels and associated increases in serum 1,25(OH)2D concentrations. A moderate increase in serum calcium concentrations was also observed that could be a consequence of the increased 1,25(OH)2D concentrations and increased intestinal calcium transport. Parathyroid hormone concentrations were diminished in the homozygous mutant mice only at 9 wk of age. Long bones displayed abnormal mineralization and a reduced growth plate. TmP/GFR was significantly increased in FGF-23 in null mutant animals.

    Conversely, transgenic mice overexpressing FGF-23 have reduced serum phosphate concentrations, increased phosphate excretion, and reduced renal sodium-phosphate cotransporter NaPi-IIa (70, 108).

    Numerous studies have now demonstrated that FGF-23 is associated with increased phosphate excretion and decreased plasma phosphate concentrations. In addition to changes in phosphate homeostasis, chronic overexpression of FGF-23 has also been linked to disturbances in vitamin D metabolism, calcium homeostasis, and increased parathyroid hormone levels (6). The exact interaction and the relative contribution of FGF-23, parathyroid hormone, and vitamin D on phosphate homeostasis in these models of chronic FGF-23 excess remain an open question.

    The mechanism of action of FGF-23 on phosphate transport is currently unknown. Limited in vitro binding studies suggest that FGF-23 may bind to FGFR-3c fusion proteins. Furthermore, tyrosine kinase inhibitors that are known to inhibit signaling through FGFRs block the effect of FGF-23 on sodium-dependent phosphate transport in opposum kidney cells (138). These results raise the possibility that FGF-23 may signal through one of the known FGFRs. Further clarification will require demonstration of FGF-23 binding to a receptor under physiologically relevant conditions.

    FGF-23 in Normal and Clinical Disorders

    Recent studies demonstrate that FGF-23 is present in normal plasma and that it is increased in a variety of circumstances associated with altered phosphate reabsorption. Elevations in circulating FGF-23 have been associated with various clinical disorders.

    Oncogenic osteomalacia. Numerous reports now show that serum FGF-23 is elevated in some, but not all, patients with oncogenic or tumor-induced osteomalacia (16, 55, 56, 72, 86, 120, 139). Removal of the tumor is generally associated with a reduction in serum FGF-23 concentrations, and there is a temporal association between the reduction in serum FGF-23 concentrations, the increase in serum phosphate concentrations, a decrease in renal phosphate wasting, and an increase in serum 1,25(OH)2D3 concentrations (56, 120, 139). In some patients, FGF-23 concentrations are not greatly increased and do not dramatically decrease following tumor removal (56). The increase in serum FGF-23 concentrations is consistent with the overexpression of mRNA and protein for FGF-23 within the tumors themselves. Recently, venous sampling has demonstrated a gradient between FGF-23 concentrations in tumor venous effluent and FGF-23 concentrations in peripheral blood, suggesting that elevations of serum FGF-23 are due to direct secretion from the tumors (120).

    Autosomal dominant hypophosphatemic rickets. As noted earlier, mutations in the FGF-23 gene within a proconvertase processing site were identified as the cause for autosomal dominant hypophosphatemic rickets (1). These mutations appear to prevent processing of FGF-23 in mice (7, 107, 134).

    X-linked hypophosphatemic rickets. The evidence for elevated FGF-23 expression in patients with X-linked hypophosphatemic rickets is conflicting. Jonsson and co-workers (56) describe elevated serum FGF-23 concentrations in many subjects, whereas, Weber and colleagues (131) reported only modestly increased or normal concentrations of FGF-23. In the Hyp mouse, preliminary reports suggest that FGF-23 serum concentrations are elevated and neutralization of FGF-23 with an antibody ameliorates hypophosphatemia and rickets (3). These data are consistent with the premise that PHEX processes FGF-23 either directly or indirectly under normal circumstances. Inactivation of PHEX, as occurs in individuals with X-linked hypophosphatemic rickets and in Hyp mice, would reduce FGF-23 degradation and cause renal phosphate wasting and hypophosphatemia. As described earlier, it is unclear whether FGF-23 is a direct substrate for PHEX (9, 16, 23, 46).

    Fibrous dysplasia. Riminucci and co-workers (96) examined the concentrations of FGF-23 in the plasma of patients with fibrous dysplasia, some of whom have hypophosphatemia. These investigators demonstrated that those patients who had low phosphate concentrations had elevated FGF-23 concentrations in the blood, whereas those who had normal phosphate concentrations did not have elevations in FGF-23 (96). These results support the association of elevated FGF-23 and hypophosphatemia.

    Chronic renal failure and hemodialysis. Serum FGF-23 concentrations are elevated in patients with chronic renal failure and in patients on hemodialysis (53, 71, 104, 137). It is attractive to hypothesize that the synthesis of FGF-23 is induced by retention of phosphate that occurs in patients at late stages of renal insufficiency. Current data demonstrating direct regulation of FGF-23 concentrations by changes in serum phosphate are conflicting. In a study by Jonsson and colleagues (71), ingestion of a diet that was either high or low in phosphate did not appreciably alter FGF-23 concentrations. A recent study suggested that serum FGF-23 concentrations are modestly increased in serum in normal human subjects receiving a high-phosphate diet; alterations in parathyroid hormone secretion were prevented by the concomitant administration of a large amount of dietary calcium (38). In this latter study, changes in FGF-23 with variations in dietary phosphate were modest, and it remains uncertain whether changes of the magnitude demonstrated in this study are sufficient to alter phosphate excretion.

    Many of the initial reports concerning elevated serum FGF-23 levels in patients with chronic renal insufficiency were obtained using an assay that recognizes both COOH-terminal fragments of the FGF-23 as well as the intact molecule. Therefore, it is difficult to assess whether the large increases in serum FGF-23 seen in these patients are due to decreased degradation of full-length FGF-23 or are FGF-23 fragments that may not have biological activity. Recent reports would suggest that the intact FGF-23 is elevated in patients with chronic renal failure (104). In many of the studies reported, a direct relationship among serum FGF-23, parathyroid hormone, phosphate, and creatinine concentrations was observed. To address the issue as to whether parathyroid hormone regulates serum FGF-23 levels, investigators have examined FGF-23 levels in patients with primary hyperparathyroidism without chronic renal failure (127, 136). There is no relationship between parathyroid hormone and FGF-23 concentrations in these patients. Furthermore, parathyroidectomy does not result in a significant alteration in FGF-23 even though parathyroid hormone concentrations decrease dramatically following surgery. Thus the stimulus driving the elevation of serum FGF-23 in chronic renal failure is not certain. Many of the uncertainties concerning the role of FGF-23 and chronic renal insufficiency would be resolved with an analysis of the different types of peptide present within the serum of such patients and a determination of the bioactivity of FGF-23 fragments. Such studies are currently under way.

    Humoral hypercalcemia of malignancy and hyperparathyroidism. Patients with humoral hypercalcemia of malignancy or primary hyperparathyroidism have elevated serum calcium concentrations as well as hypophosphatemia. It is possible that increased circulating FGF-23 could contribute to the hypophosphatemia seen in individuals with these disorders. Recently, serum FGF-23 has been shown to be elevated in patients with humoral hypercalcemia of malignancy (127). The concentrations are elevated 5- to 10-fold. Interestingly, the elevations in FGF-23 are not correlated with the concentrations of serum phosphorus. The finding of elevated FGF-23 concentrations in patients with humoral hypercalcemia of malignancy may explain the reduced 1,25-dihydroxyvitamin D concentrations seen in these patients (118). Patients with primary hyperparathyroidism have slightly elevated FGF-23 concentrations that do not change after parathyroidectomy (127).

    Ovarian cancer. Because FGF physiology is known to be altered in patients with ovarian cancer, we measured FGF-23 concentrations in patients with early and late ovarian cancer (126). Interestingly, we found that patients with stage three and four ovarian cancer had elevated FGF-23 concentrations when measured by both COOH-terminal and intact FGF-23 immunoassays. It is interesting to speculate that FGF-23 may be a tumor marker as it is increased in both ovarian cancers and in patients with humoral hypercalcemia of malignancy. In these circumstances, FGF-23 appears not to correlate with phosphate concentrations and might indicate that FGF-23 concentrations need to be significantly elevated to induce hypophosphatemia, or that other factors are involved in the pathogenesis of low phosphate concentrations and renal phosphate wasting.

    Tumoral calcinosis. Tumoral calcinosis is an unusual disorder associated with hyperphosphatemia, elevated 1,25(OH)2D3 concentrations, and reduced excretion of phosphorus in the urine. This disorder has recently been shown to be due to a defect in the activity of an O-linked glycosylating enzyme, GALNT3 (128). Interestingly, serum FGF-23 concentrations are high in these patients. It is uncertain from the published report whether the serum FGF-23 concentrations were measured using an assay that detects intact FGF-23 or FGF-23 fragments as well as the intact molecule. It is possible that there could be defects in the processing of FGF-23 as a result of a failure in glycosylation of the molecule, or as a result of the chronic hypophosphatemia seen in these patients. A recent report suggests that the hyperostosis-hyperphosphatemia syndrome is an allelic disorder similar to tumoral calcinosis (40). No reports of FGF-23 concentrations in this syndrome are available.

    BIOLOGICAL PROPERTIES OF sFRP-4

    sFRP-4 was among the most consistently overexpressed genes found associated with oncogenic osteomalacia (Table 2). To assess whether sFRP-4 has phosphatonin activity, it was expressed by recombinant methods in COS or insect cells (10). Increasing concentrations of the recombinant protein were added to opossum kidney cells to determine whether it inhibits sodium-dependent phosphate transport. We observed that sFRP-4 inhibited sodium-dependent phosphate transport in opossum kidney cells in a dose-dependent manner at concentrations in the picograms per milliliter range (Fig. 5A). When infused into rats, sFRP-4 increased renal phosphate excretion at both 2 and 8 h following initiation of the sFRP-4 infusion (10) (Figs. 5C and 6A). Minimal changes in sodium excretion were seen, and calcium excretion did not change. Interestingly, the effects of sFRP-4 were also demonstrated in parathyroidectomized rats, thus demonstrating that parathyroid hormone was not essential for the phosphaturic effect of sFRP-4 (Fig. 6B). During an 8-h intravenous infusion of sFRP-4, serum phosphate concentrations decreased and phosphate excretion increased. However, no change in 25-hydroxyvitamin D 1-hydroxylase mRNA concentrations was noted in the kidney. The infusion of sFRP-4 was associated with a decrease in -catenin concentrations in renal cells and an increase in phosphorylated -catenin, thereby demonstrating that sFRP-4 may act as an antagonist against Wnt molecules in the kidney (Fig. 6C). Additionally, sFRP-4 was detected in the plasma of patients with tumor-induced osteomalacia, although elevated levels were not found with the current assay. Thus the data published to date suggest that sFRP-4 is a phosphatonin. Details concerning the mechanism by which FRP-4s inhibits renal phosphate reabsorption and its relationship to FGF-23 will need to be elucidated in the future.

    BIOLOGICAL PROPERTIES OF MEPE

    MEPE is also among the most abundantly overexpressed mRNA species found in tumors associated with renal phosphate wasting and osteomalacia (29, 98, 100). Recently, MEPE has been expressed in insects cells and administered to mice in vivo (100). The protein causes renal phosphate wasting and a reduction in serum phosphate concentrations in vivo. Additionally, inhibition of sodium-dependent phosphate uptake was noted in opossum kidney cells exposed to the protein. MEPE also appears to inhibit bone mineralization in vitro, and MEPE null mice have increased bone mineralization (42). This suggests that it may play a role in the pathogenesis of X-linked hypophosphatemic rickets, in which there is phosphate wasting and evidence for a mineralization defect that is independent of low phosphate concentrations in the extracellular fluid (135). Recent evidence suggests that concentrations of this substance is increased in the serum of patients with X-linked hypophosphatemic rickets (18). It has been suggested that MEPE is a substrate for PHEX (47, 99) and that PHEX prevents proteolysis of MEPE and release of a protease-resistant MEPE-ASARM peptide, an inhibitor of mineralization (minhibin). Phex may be acting to interfere with the actions of other enzymes that degrade extracellular matrix proteins. PHEX and MEPE form a nonproteolytic protein interaction via the MEPE COOH-terminal ASARM motif. The ASARM peptide is believed to inhibit mineralization in vivo. The binding of MEPE and ASARM peptide by PHEX may explain why loss of functional osteoblast-expressed PHEX results in defective mineralization in Hyp. MEPE concentrations have been measured in normal humans, and concentrations of the protein appear to correlate with bone mineral density and serum phosphate concentrations (54).

    FGF-7 IS OVEREXPRESSED IN TUMORS ASSOCIATED WITH OSTEOMALACIA AND RENAL PHOSPHATE WASTING

    A recent report has shown that FGF-7 is overexpressed in tumors associated with osteomalacia and renal phosphate wasting (24). FGF-7 protein inhibited sodium-dependent phosphate transport in opossum kidney cells. Anti-FGF-7 antibodies attenuated the inhibitory effect of tumor supernatants on sodium-dependent phosphate transport. Only low concentrations of FGF-23 were present in the conditioned medium of tumor cells. At present it is not known whether FGF-7 circulates in plasma, whether it alters 25 hydroxyvitamin D 1-hydroxylase levels, or whether it is elevated in the plasma of subjects with tumor-induced osteomalacia. Nevertheless, the report does point to the complexity of factors involved in the pathogenesis of tumor-induced osteomalacia.

    UNANSWERED QUESTIONS AND FUTURE DIRECTIONS FOR RESEARCH

    Several new phosphaturic peptides have now been isolated from tumors associated with renal phosphate wasting and osteomalacia. The most information concerning these various peptides is available for FGF-23. Concentrations of this protein are elevated in most patients with tumor-induced osteomalacia and are reduced following removal of the tumor. Of interest, however, is the observation that FGF-23 concentrations are elevated in patients who do not have oncogenic osteomalacia but have tumors associated with humoral hypercalcemia of malignancy. Another report shows that FGF-23 concentrations are elevated in patients with malignancies not associated with phosphate wasting. Hence, it is possible that FGF-23 concentrations must be significantly elevated before the occurrence of hypophosphatemia or that other factors might also need to be formed in excessive amounts to cause hypophosphatemia in tumor-induced osteomalacia.

    Another key, and unresolved question, relates to whether these peptides play a role in the control of phosphate homeostasis under normal circumstances. It is attractive to consider that FGF-23 may play a role in the control of renal phosphate reabsorption under normal circumstances. Studies performed in humans, however, have shown either no or only modest changes in FGF-23 concentrations following the alteration of dietary phosphate. Preliminary data in rodents suggest that serum FGF-23 may be significantly altered by dietary phosphate (89). Thus it is still unclear whether changes in serum FGF-23 may be responsible for alterations in phosphate excretion in response to changes in serum phosphate levels.

    Less information is available concerning role of sFRP-4 and MEPE in various clinical disorders. In part, this relates to the fact that robust assays that distinguish modified forms such as proteolytic fragments derived from the intact molecule are not available for these proteins. Furthermore, analysis of FRP-4 and MEPE expression in various clinical conditions is just beginning.

    The question as to why there are multiple phosphatonin molecules needs to be resolved in the future. It is possible that one or the other molecule acts downstream of the other. For example, the administration of FGF-23 may be associated with an induction of sFRP-4 in renal cells. FGF-23 may have similar effects on MEPE in bone cells. Suffice it to say that study of rare diseases associated with phosphate wasting or abnormal phosphate retention has provided new insights into the mechanisms by which phosphate homeostasis is regulated.

    GRANTS

    This study was supported by National Institute of Diabetes and Digestive and Kidney Diseases Grants DK-25409, DK-58546, and DK-65830 and a grant from the Genzyme Corporation.

    FOOTNOTES

    REFERENCES

    ADHR Consortium Autosomal.dominant hypophosphataemic rickets is associated with mutations in FGF23. Nat Genet 26: 345–348, 2000.

    Anonymous. A gene (PEX) with homologies to endopeptidases is mutated in patients with X.-linked hypophosphatemic rickets. The HYP Consortium. Nat Genet 11: 130–136, 1995.

    Aono Y, Shimada T, Yamazaki Y, Hino R, Takeuchi Y, Fujita T, Fukumoto S, Nagano N, Wada M, and Yamashita T. The neutralization of FGF-23 ameliorates hypophosphatemia and rickets in Hyp mice (Abstract). J Bone Miner Res 18: S16, 2003.

    Aurbach GD and Chase LR. Cyclic 3',5'-adenylic acid in bone and the mechanism of action of parathyroid hormone. Federation Proc 29: 1179–1182, 1970.

    Bacic D, Wagner CA, Hernando N, Kaissling B, Biber J, and Murer H. Novel aspects in regulated expression of the renal type IIa Na/Pi-cotransporter. Kidney Int Suppl: S5–S12, 2004.

    Bai X, Miao D, Li J, Goltzman D, and Karaplis AC. Transgenic mice overexpressing human fibroblast growth factor 23 (R176Q) delineate a putative role for parathyroid hormone in renal phosphate wasting disorders. Endocrinology 145: 5269–5279, 2004.

    Bai XY, Miao D, Goltzman D, and Karaplis AC. The autosomal dominant hypophosphatemic rickets R176Q mutation in fibroblast growth factor 23 resists proteolytic cleavage and enhances in vivo biological potency. J Biol Chem 278: 9843–9849, 2003.

    Baxter LA and DeLuca HF. Stimulation of 25-hydroxyvitamin D3-1-hydroxylase by phosphate depletion. J Biol Chem 251: 3158–3161, 1976.

    Benet-Pages A, Lorenz-Depiereux B, Zischka H, White KE, Econs MJ, and Strom TM. FGF23 is processed by proprotein convertases but not by PHEX. Bone 35: 455–462, 2004.

    Berndt T, Craig TA, Bowe AE, Vassiliadis J, Reczek D, Finnegan R, Jan De Beur SM, Schiavi SC, and Kumar R. Secreted frizzled-related protein 4 is a potent tumor-derived phosphaturic agent. J Clin Invest 112: 785–794, 2003.

    Berndt T and Knox F. Renal regulation of phosphate excretion. In: The Kidney: Physiology and Pathophysiology, edited by Seldin DW and Giebisch G. New York: Raven, 1992, p. 2511–2532.

    Biber J, Custer M, Magagnin S, Hayes G, Werner A, Lotscher M, Kaissling B, and Murer H. Renal Na/Pi-cotransporters. Kidney Int 49: 981–985, 1996.

    Biber J, Custer M, Werner A, Kaissling B, and Murer H. Localization of NaPi-1, a Na/Pi cotransporter, in rabbit kidney proximal tubules. II. Localization by immunohistochemistry. Pflügers Arch 424: 210–215, 1993.

    Biber J and Murer H. A molecular view of renal Na-dependent phosphate transport. Renal Physiol Biochem 17: 212–215, 1994.

    Biber J, Murer H, and Forster I. The renal type II Na+/phosphate cotransporter. J Bioenerg Biomembr 30: 187–194, 1998.

    Bowe AE, Finnegan R, Jan de Beur SM, Cho J, Levine MA, Kumar R, and Schiavi SC. FGF-23 inhibits renal tubular phosphate transport and is a PHEX substrate. Biochem Biophys Res Commun 284: 977–981, 2001.

    Boyle IT, Gray RW, and DeLuca HF. Regulation by calcium of in vivo synthesis of 1,25-dihydroxycholecalciferol and 21,25-dihydroxycholecalciferol. Proc Natl Acad Sci USA 68: 2131–2134, 1971.

    Bresler D, Bruder J, Mohnike K, Fraser WD, and Rowe PS. Serum MEPE-ASARM-peptides are elevated in X-linked rickets (HYP): implications for phosphaturia and rickets. J Endocrinol 183: R1–R9, 2004.

    Bricker NS. On the pathogenesis of the uremic state. An exposition of the "trade-off hypothesis." N Engl J Med 286: 1093–1099, 1972.

    Bricker NS. On the pathogenesis of the uremic state—an exposition of the trade-off hypothesis. Nippon Jinzo Gakkai Shi 16: 327–339, 1974.

    Bricker NS, Bourgoignie J, Weber H, Schmidt RW, and Slatopolsky E. Pathogenesis of the uremic state: a new perspective. Adv Nephrol Necker Hosp 2: 263–276, 1972.

    Cai Q, Hodgson SF, Kao PC, Lennon VA, Klee GG, Zinsmiester AR, and Kumar R. Brief report: inhibition of renal phosphate transport by a tumor product in a patient with oncogenic osteomalacia. N Engl J Med 330: 1645–1649, 1994.

    Campos M, Couture C, Hirata IY, Juliano MA, Loisel TP, Crine P, Juliano L, Boileau G, and Carmona AK. Human recombinant endopeptidase PHEX has a strict S1' specificity for acidic residues and cleaves peptides derived from fibroblast growth factor-23 and matrix extracellular phosphoglycoprotein. Biochem J 373: 271–279, 2003.

    Carpenter TO, Ellis BK, Insogna KL, Philbrick WM, Sterpka J, and Shimkets R. Fibroblast growth factor 7: an inhibitor of phosphate transport derived from oncogenic osteomalacia-causing tumors. J Clin Endocrinol Metab 90: 1012–1020, 2004.

    Castillo L, Tanaka Y, and DeLuca HF. The mobilization of bone mineral by 1,25-dihydroxyvitamin D3 in hypophosphatemic rats. Endocrinology 97: 995–999, 1975.

    Chase LR and Aurbach GD. Parathyroid function and the renal excretion of 3'5'-adenylic acid. Proc Natl Acad Sci USA 58: 518–525, 1967.

    Cohen P. The structure and regulation of protein phosphatases. Annu Rev Biochem 58: 453–508, 1989.

    Custer M, Meier F, Schlatter E, Greger R, Garcia-Perez A, Biber J, and Murer H. Localization of NaPi-1, a Na-Pi cotransporter, in rabbit kidney proximal tubules. I. mRNA localization by reverse transcription/polymerase chain reaction. Pflügers Arch 424: 203–209, 1993.

    De Beur SM, Finnegan RB, Vassiliadis J, Cook B, Barberio D, Estes S, Manavalan P, Petroziello J, Madden SL, Cho JY, Kumar R, Levine MA, and Schiavi SC. Tumors associated with oncogenic osteomalacia express genes important in bone and mineral metabolism. J Bone Miner Res 17: 1102–1110, 2002.

    DeLuca HF. The kidney as an endocrine organ for the production of 1,25-dihydroxyvitamin D3, a calcium-mobilizing hormone. N Engl J Med 289: 359–365, 1973.

    Dominguez JH, Gray RW, and Lemann J Jr. Dietary phosphate deprivation in women and men: effects on mineral and acid balances, parathyroid hormone and the metabolism of 25-OH-vitamin D. J Clin Endocrinol Metab 43: 1056–1068, 1976.

    Drezner MK. Hypophosphatemic rickets. Endocr Dev 6: 126–155, 2003.

    Drezner MK. The role of abnormal vitamin D metabolism in X-linked hypophosphatemic rickets and osteomalacia. Adv Exp Med Biol 178: 399–404, 1984.

    Du L, Desbarats M, Viel J, Glorieux FH, Cawthorn C, and Ecarot B. cDNA cloning of the murine Pex gene implicated in X-linked hypophosphatemia and evidence for expression in bone. Genomics 36: 22–28, 1996.

    Econs MJ and Drezner MK. Tumor-induced osteomalacia—unveiling a new hormone. N Engl J Med 330: 1679–1681, 1994.

    Econs MJ, Feussner JR, Samsa GP, Effman EL, Vogler JB, Martinez S, Friedman NE, Quarles LD, and Drezner MK. X-linked hypophosphatemic rickets without "rickets". Skeletal Radiol 20: 109–114, 1991.

    Econs MJ, Samsa GP, Monger M, Drezner MK, and Feussner JR. X-linked hypophosphatemic rickets: a disease often unknown to affected patients. Bone Miner 24: 17–24, 1994.

    Ferrari SL, Bonjour JP, and Rizzoli R. Fibroblast growth factor-23 relationship to dietary phosphate and renal phosphate handling in healthy young men. J Clin Endocrinol Metab 9: 1519–1524, 2004.

    Fleisch H. Homeostasis of inorganic phosphate. In: Fundamental and Clinical Bone Physiology, edited by Urist MR. Philadelphia, PA: Lippincott, 1980, p. 268–282.

    Frishberg Y, Topaz O, Bergman R, Behar D, Fisher D, Gordon D, Richard G, and Sprecher E. Identification of a recurrent mutation in GALNT3 demonstrates that hyperostosis-hyperphosphatemia syndrome and familial tumoral calcinosis are allelic disorders. J Mol Med 83: 33–38, 2005.

    Garabedian M, Holick MF, Deluca HF, and Boyle IT. Control of 25-hydroxycholecalciferol metabolism by parathyroid glands. Proc Natl Acad Sci USA 69: 1673–1676, 1972.

    Gowen LC, Petersen DN, Mansolf AL, Qi H, Stock JL, Tkalcevic GT, Simmons HA, Crawford DT, Chidsey-Frink KL, Ke HZ, McNeish JD, and Brown TA. Targeted disruption of the osteoblast/osteocyte factor 45 gene (OF45) results in increased bone formation and bone mass. J Biol Chem 278: 1998–2007, 2003.

    Gray RW. Control of plasma 1,25-(OH)2-vitamin D concentrations by calcium and phosphorus in the rat: effects of hypophysectomy. Calcif Tissue Int 33: 485–488, 1981.

    Gray RW. Effects of age and sex on the regulation of plasma 1,25-(OH)2-D by phosphorus in the rat. Calcif Tissue Int 33: 477–484, 1981.

    Gray RW, Haasch ML, and Brown CE. Regulation of plasma 1,25-(OH)2-D3 by phosphate: evidence against a role for total or acid-soluble renal phosphate content. Calcif Tissue Int 35: 773–777, 1983.

    Guo R, Liu S, Spurney RF, and Quarles LD. Analysis of recombinant Phex: an endopeptidase in search of a substrate. Am J Physiol Endocrinol Metab 281: E837–E847, 2001.

    Guo R, Rowe PS, Liu S, Simpson LG, Xiao ZS, and Darryl Quarles LD. Inhibition of MEPE cleavage by Phex. Biochem Biophys Res Commun 297: 38–45, 2002.

    Hattenhauer O, Traebert M, Murer H, and Biber J. Regulation of small intestinal Na-Pi type IIb cotransporter by dietary phosphate intake. Am J Physiol Gastrointest Liver Physiol 277: G756–G762, 1999.

    Hernando N, Forgo J, Biber J, and Murer H. PTH-Induced downregulation of the type IIa Na/Pi-cotransporter is independent of known endocytic motifs. J Am Soc Nephrol 11: 1961–1968, 2000.

    Holick MF and DeLuca HF. Vitamin D metabolism. Annu Rev Med 25: 349–367, 1974.

    Hubbard SR and Till JH. Protein tyrosine kinase structure and function. Annu Rev Biochem 69: 373–398, 2000.

    Hunter T and Cooper JA. Protein-tyrosine kinases. Annu Rev Biochem 54: 897–930, 1985.

    Imanishi Y, Inaba M, Nakatsuka K, Nagasue K, Okuno S, Yoshihara A, Miura M, Miyauchi A, Kobayashi K, Miki T, Shoji T, Ishimura E, and Nishizawa Y. FGF-23 in patients with end-stage renal disease on hemodialysis. Kidney Int 65: 1943–1946, 2004.

    Jain A, Fedarko NS, Collins MT, Gelman R, Ankrom MA, Tayback M, and Fisher LW. Serum levels of matrix extracellular phosphoglycoprotein (MEPE) in normal humans correlate with serum phosphorus, parathyroid hormone and bone mineral density. J Clin Endocrinol Metab 89: 4158–4161, 2004.

    John MR, Wickert H, Zaar K, Jonsson KB, Grauer A, Ruppersberger P, Schmidt-Gayk H, Murer H, Ziegler R, and Blind E. A case of neuroendocrine oncogenic osteomalacia associated with a PHEX and fibroblast growth factor-23 expressing sinusoidal malignant schwannoma. Bone 29: 393–402, 2001.

    Jonsson KB, Zahradnik R, Larsson T, White KE, Sugimoto T, Imanishi Y, Yamamoto T, Hampson G, Koshiyama H, Ljunggren O, Oba K, Yang IM, Miyauchi A, Econs MJ, Lavigne J, and Juppner H. Fibroblast growth factor 23 in oncogenic osteomalacia and X-linked hypophosphatemia. N Engl J Med 348: 1656–1663, 2003.

    Kaplan MA, Canterbury JM, Gavellas G, Jaffe D, Bourgoignie JJ, Reiss E, and Bricker NS. Interrelations between phosphorus, calcium, parathyroid hormone, and renal phosphate excretion in response to an oral phosphorus load in normal and uremic dogs. Kidney Int 14: 207–214, 1978.

    Kaplan MA, Canterbury JM, Gavellas G, Reiss E, and Bricker NS. Interrelations between phosphorus, calcium, parathyroid hormone, and phosphate excretion in the normal and uremic dog. Adv Exp Med Biol 103: 257–262, 1978.

    Karim-Jimenez Z, Hernando N, Biber J, and Murer H. A dibasic motif involved in parathyroid hormone-induced down-regulation of the type IIa NaPi cotransporter. Proc Natl Acad Sci USA 97: 12896–12901, 2000.

    Keusch I, Traebert M, Lotscher M, Kaissling B, Murer H, and Biber J. Parathyroid hormone and dietary phosphate provoke a lysosomal routing of the proximal tubular Na/Pi-cotransporter type II. Kidney Int 54: 1224–1232, 1998.

    Knochel JP. The pathophysiology and clinical characteristics of severe hypophosphatemia. Arch Intern Med 137: 203–220, 1977.

    Knochel JP, Barcenas C, Cotton JR, Fuller TJ, Haller R, and Carter NW. Hypophosphatemia and rhabdomyolysis. Trans Assoc Am Physicians 91: 156–168, 1978.

    Krebs EG and Beavo JA. Phosphorylation-dephosphorylation of enzymes. Annu Rev Biochem 48: 923–959, 1979.

    Kumar R. The metabolism of 1,25-dihydroxyvitamin D3. Endocr Rev 1: 258–267, 1980.

    Kumar R. Metabolism of 1,25-dihydroxyvitamin D3. Physiol Rev 64: 478–504, 1984.

    Kumar R. Phosphatonin—a new phosphaturetic hormone (lessons from tumour-induced osteomalacia and X-linked hypophosphataemia). Nephrol Dial Transplant 12: 11–13, 1997.

    Kumar R. Tumor-induced osteomalacia and the regulation of phosphate homeostasis. Bone 27: 333–338, 2000.

    Kumar R and Riggs B. Pathologic bone physiology. In: Fundamental and Clinical Bone Physiology, edited by Urist MR. Philadelphia, PA: Lippincott, 1980, p. 394–406.

    Lambert G, Traebert M, Hernando N, Biber J, and Murer H. Studies on the topology of the renal type II NaPi-cotransporter. Pflügers Arch 437: 972–978, 1999.

    Larsson T, Marsell R, Schipani E, Ohlsson C, Ljunggren O, Tenenhouse HS, Juppner H, and Jonsson KB. Transgenic mice expressing fibroblast growth factor 23 under the control of the (I) collagen promoter exhibit growth retardation, osteomalacia, and disturbed phosphate homeostasis. Endocrinology 145: 3087–3094, 2004.

    Larsson T, Nisbeth U, Ljunggren O, Juppner H, and Jonsson KB. Circulating concentration of FGF-23 increases as renal function declines in patients with chronic kidney disease, but does not change in response to variation in phosphate intake in healthy volunteers. Kidney Int 64: 2272–2279, 2003.

    Larsson T, Zahradnik R, Lavigne J, Ljunggren O, Juppner H, and Jonsson KB. Immunohistochemical detection of FGF-23 protein in tumors that cause oncogenic osteomalacia. Eur J Endocrinol 148: 269–276, 2003.

    Meyer RA Jr, Henley CM, Meyer MH, Morgan PL, McDonald AG, Mills C, and Price DK. Partial deletion of both the spermine synthase gene and the Pex gene in the X-linked hypophosphatemic, gyro (Gy) mouse. Genomics 48: 289–295, 1998.

    Meyer RA Jr, Meyer MH, and Gray RW. Parabiosis suggests a humoral factor is involved in X-linked hypophosphatemia in mice. J Bone Miner Res 4: 493–500, 1989.

    Meyer RA Jr, Tenenhouse HS, Meyer MH, and Klugerman AH. The renal phosphate transport defect in normal mice parabiosed to X-linked hypophosphatemic mice persists after parathyroidectomy. J Bone Miner Res 4: 523–532, 1989.

    Murer H. Cellular mechanisms in proximal tubular Pi reabsorption: some answers and more questions. J Am Soc Nephrol 2: 1649–1665, 1992.

    Murer H and Biber J. Control of proximal tubular apical Na/Pi cotransport. Exp Nephrol 4: 201–204, 1996.

    Murer H and Biber J. Membrane traffic and control of proximal tubular sodium phosphate (Na/Pi)-cotransport. Wien Klin Wochenschr 109: 441–444, 1997.

    Murer H and Biber J. Molecular mechanisms of renal apical Na/phosphate cotransport. Annu Rev Physiol 58: 607–618, 1996.

    Murer H and Biber J. Renal sodium-phosphate cotransport. Curr Opin Nephrol Hypertens 3: 504–510, 1994.

    Murer H and Biber J. Renal tubular phosphate transport. In: The Kidney: Physiology and Pathophysiology, edited by Seldin DW and Giebisch G. New York: Raven, 1992, p. 2481–2509.

    Murer H, Forster I, Hilfiker H, Pfister M, Kaissling B, Lotscher M, and Biber J. Cellular/molecular control of renal Na/Pi-cotransport. Kidney Int Suppl 65: S2–S10, 1998.

    Murer H, Hernando N, Forster I, and Biber J. Molecular aspects in the regulation of renal inorganic phosphate reabsorption: the type IIa sodium/inorganic phosphate co-transporter as the key player. Curr Opin Nephrol Hypertens 10: 555–561, 2001.

    Murer H, Hernando N, Forster I, and Biber J. Proximal tubular phosphate reabsorption: molecular mechanisms. Physiol Rev 80: 1373–1409, 2000.

    Murer H, Lotscher M, Kaissling B, and Biber J. Molecular mechanisms in the regulation of renal proximal tubular Na/phosphate cotransport. Kidney Blood Press Res 19: 151–154, 1996.

    Nelson AE, Bligh RC, Mirams M, Gill A, Au A, Clarkson A, Juppner H, Ruff S, Stalley P, Scolyer RA, Robinson BG, Mason RS, and Bligh PC. Clinical case seminar: fibroblast growth factor 23: a new clinical marker for oncogenic osteomalacia. J Clin Endocrinol Metab 88: 4088–4094, 2003.

    Nesbitt T, Coffman TM, Griffiths R, and Drezner MK. Crosstransplantation of kidneys in normal and Hyp mice. Evidence that the Hyp mouse phenotype is unrelated to an intrinsic renal defect. J Clin Invest 89: 1453–1459, 1992.

    Neuman W. Bone material and calcification mechanisms. In: Fundamental and Clinical Bone Physiology, edited by Urist MR. Philadelphia, PA: Lippincott, 1980, p. 83–107.

    Perwad F, Azam M, Zhang M, Yamashita T, Tenenhouse H, and Portale A. Dietary phosphorus regulates serum FGF-23 concentrations and 1, 25(OH)2D3 metabolism in mice (Abstract). J Bone Miner Res 19: S251, 2004.

    Pfister MF, Forgo J, Ziegler U, Biber J, and Murer H. cAMP-dependent and -independent downregulation of type II Na-Pi cotransporters by PTH. Am J Physiol Renal Physiol 276: F720–F725, 1999.

    Pfister MF, Lederer E, Forgo J, Ziegler U, Lotscher M, Quabius ES, Biber J, and Murer H. Parathyroid hormone-dependent degradation of type II Na+/Pi cotransporters. J Biol Chem 272: 20125–20130, 1997.

    Pfister MF, Ruf I, Stange G, Ziegler U, Lederer E, Biber J, and Murer H. Parathyroid hormone leads to the lysosomal degradation of the renal type II Na/Pi cotransporter. Proc Natl Acad Sci USA 95: 1909–1914, 1998.

    Polisson RP, Martinez S, Khoury M, Harrell RM, Lyles KW, Friedman N, Harrelson JM, Reisner E, and Drezner MK. Calcification of entheses associated with X-linked hypophosphatemic osteomalacia. N Engl J Med 313: 1–6, 1985.

    Posillico JT, Lobaugh B, Muhlbaier LH, and Drezner MK. Abnormal parathyroid function in the X-linked hypophosphatemic mouse. Calcif Tissue Int 37: 418–422, 1985.

    Potts JT Jr, Tregear GW, Keutmann HT, Niall HD, Sauer R, Deftos LJ, Dawson BF, Hogan ML, and Aurbach GD. Synthesis of a biologically active N-terminal tetratriacontapeptide of parathyroid hormone. Proc Natl Acad Sci USA 68: 63–67, 1971.

    Riminucci M, Collins MT, Fedarko NS, Cherman N, Corsi A, White KE, Waguespack S, Gupta A, Hannon T, Econs MJ, Bianco P, and Gehron Robey P. FGF-23 in fibrous dysplasia of bone and its relationship to renal phosphate wasting. J Clin Invest 112: 683–692, 2003.

    Rizzoli R, Caverzasio J, Fleisch H, and Bonjour JP. Parathyroid hormone-like changes in renal calcium and phosphate reabsorption induced by Leydig cell tumor in thyroparathyroidectomized rats. Endocrinology 119: 1004–1009, 1986.

    Rowe PS, de Zoysa PA, Dong R, Wang HR, White KE, Econs MJ, and Oudet CL. MEPE, a new gene expressed in bone marrow and tumors causing osteomalacia. Genomics 67: 54–68, 2000.

    Rowe PS, Garrett IR, Schwarz PM, Carnes DL, Lafer EM, Mundy GR, and Gutierrez GE. Surface plasmon resonance (SPR) confirms that MEPE binds to PHEX via the MEPE-ASARM motif: a model for impaired mineralization in X-linked rickets (HYP). Bone 36: 33–46, 2005.

    Rowe PS, Kumagai Y, Gutierrez G, Garrett IR, Blacher R, Rosen D, Cundy J, Navvab S, Chen D, Drezner MK, Quarles LD, and Mundy GR. MEPE has the properties of an osteoblastic phosphatonin and minhibin. Bone 34: 303–319, 2004.

    Schiavi SC and Kumar R. The phosphatonin pathway: new insights in phosphate homeostasis. Kidney Int 65: 1–14, 2004.

    Schiavi SC and Moe OW. Phosphatonins: a new class of phosphate-regulating proteins. Curr Opin Nephrol Hypertens 11: 423–430, 2002.

    Segawa H, Kaneko I, Yamanaka S, Ito M, Kuwahata M, Inoue Y, Kato S, and Miyamoto K. Intestinal Na-Pi cotransporter adaptation to dietary Pi content in vitamin D receptor null mice. Am J Physiol Renal Physiol 287: F39–F47, 2004.

    Shigematsu T, Kazama JJ, Yamashita T, Fukumoto S, Hosoya T, Gejyo F, and Fukagawa M. Possible involvement of circulating fibroblast growth factor 23 in the development of secondary hyperparathyroidism associated with renal insufficiency. Am J Kidney Dis 44: 250–256, 2004.

    Shimada T, Kakitani M, Yamazaki Y, Hasegawa H, Takeuchi Y, Fujita T, Fukumoto S, Tomizuka K, and Yamashita T. Targeted ablation of Fgf23 demonstrates an essential physiological role of FGF23 in phosphate and vitamin D metabolism. J Clin Invest 113: 561–568, 2004.

    Shimada T, Mizutani S, Muto T, Yoneya T, Hino R, Takeda S, Takeuchi Y, Fujita T, Fukumoto S, and Yamashita T. Cloning and characterization of FGF23 as a causative factor of tumor-induced osteomalacia. Proc Natl Acad Sci USA 98: 6500–6505, 2001.

    Shimada T, Muto T, Urakawa I, Yoneya T, Yamazaki Y, Okawa K, Takeuchi Y, Fujita T, Fukumoto S, and Yamashita T. Mutant FGF-23 responsible for autosomal dominant hypophosphatemic rickets is resistant to proteolytic cleavage and causes hypophosphatemia in vivo. Endocrinology 143: 3179–3182, 2002.

    Shimada T, Urakawa I, Yamazaki Y, Hasegawa H, Hino R, Yoneya T, Takeuchi Y, Fujita T, Fukumoto S, and Yamashita T. FGF-23 transgenic mice demonstrate hypophosphatemic rickets with reduced expression of sodium phosphate cotransporter type IIa. Biochem Biophys Res Commun 314: 409–414, 2004.

    Slatopolsky E and Bricker NS. The role of phosphorus restriction in the prevention of secondary hyperparathyroidism in chronic renal disease. Kidney Int 4: 141–145, 1973.

    Slatopolsky E, Caglar S, Gradowska L, Canterbury J, Reiss E, and Bricker NS. On the prevention of secondary hyperparathyroidism in experimental chronic renal disease using "proportional reduction" of dietary phosphorus intake. Kidney Int 2: 147–151, 1972.

    Slatopolsky E, Gradowska L, Kashemsant C, Keltner R, Manley C, and Bricker NS. The control of phosphate excretion in uremia. J Clin Invest 45: 672–677, 1966.

    Slatopolsky E, Robson AM, Elkan I, and Bricker NS. Control of phosphate excretion in uremic man. J Clin Invest 47: 1865–1874, 1968.

    Sorribas V, Markovich D, Hayes G, Stange G, Forgo J, Biber J, and Murer H. Cloning of a Na/Pi cotransporter from opossum kidney cells. J Biol Chem 269: 6615–6621, 1994.

    Steele TH. Effect of phosphorus depletion on the renal transport of phosphate. Adv Exp Med Biol 103: 343–355, 1978.

    Steele TH. Interactions of starvation and selective phosphorus depletion on renal phosphate reabsorption. Renal Physiol 5: 44–52, 1982.

    Steele TH, Engle JE, Tanaka Y, Lorenc RS, Dudgeon KL, and DeLuca HF. Phosphatemic action of 1,25-dihydroxyvitamin D3. Am J Physiol 229: 489–495, 1975.

    Steele TH and Underwood JL. Renal response to phosphorus deprivation in the isolated rat kidney. Kidney Int 13: 124–128, 1978.

    Stewart AF, Horst R, Deftos LJ, Cadman EC, Lang R, and Broadus AE. Biochemical evaluation of patients with cancer-associated hypercalcemia: evidence for humoral and nonhumoral groups. N Engl J Med 303: 1377–1383, 1980.

    Strewler GJ. FGF23, hypophosphatemia, and rickets: has phosphatonin been found Proc Natl Acad Sci USA 98: 5945–5946, 2001.

    Takeuchi Y, Suzuki H, Ogura S, Imai R, Yamazaki Y, Yamashita T, Miyamoto Y, Okazaki H, Nakamura K, Nakahara K, Fukumoto S, and Fujita T. Venous sampling for fibroblast growth factor-23 confirms preoperative diagnosis of tumor-induced osteomalacia. J Clin Endocrinol Metab 89: 3979–3982, 2004.

    Tanaka Y and DeLuca HF. The control of 25-hydroxyvitamin D metabolism by inorganic phosphorus. Arch Biochem Biophys 154: 566–574, 1973.

    Tanaka Y and DeLuca HF. Role of 1,25-dihydroxyvitamin D3 in maintaining serum phosphorus and curing rickets. Proc Natl Acad Sci USA 71: 1040–1044, 1974.

    Tanaka Y, Frank H, and DeLuca HF. Biological activity of 1,25-dihydroxyvitamin D3 in the rat. Endocrinology 92: 417–422, 1973.

    Tanaka Y, Frank H, and DeLuca HF. Intestinal calcium transport: stimulation by low phosphorus diets. Science 181: 564–566, 1973.

    Tanaka Y, Frank H, and DeLuca HF. Role of 1,25-dihydroxycholecalciferol in calcification of bone and maintenance of serum calcium concentration in the rat. J Nutr 102: 1569–1577, 1972.

    Tebben P, Kalli K, Cliby W, Hartmann L, Grande J, Singh RJ, and Kumar R. Elevated fibroblast growth factor 23 concentrations in women with malignant ovarian tumors. Mayo Clin Proc 80: 745–751, 2005.

    Tebben PJ, Singh RJ, Clarke BL, and Kumar R. Fibroblast growth factor 23, parathyroid hormone, and 1,25-dihydroxyvitamin D in surgically treated primary hyperparathyroidism. Mayo Clin Proc 79: 1508–1513, 2004.

    Topaz O, Shurman DL, Bergman R, Indelman M, Ratajczak P, Mizrachi M, Khamaysi Z, Behar D, Petronius D, Friedman V, Zelikovic I, Raimer S, Metzker A, Richard G, and Sprecher E. Mutations in GALNT3, encoding a protein involved in O-linked glycosylation, cause familial tumoral calcinosis. Nat Genet 36: 579–581, 2004.

    Traebert M, Roth J, Biber J, Murer H, and Kaissling B. Internalization of proximal tubular type II Na-Pi cotransporter by PTH: immunogold electron microscopy. Am J Physiol Renal Physiol 278: F148–F154, 2000.

    Traebert M, Volkl H, Biber J, Murer H, and Kaissling B. Luminal and contraluminal action of 1–34 and 3–34 PTH peptides on renal type IIa Na-Pi cotransporter. Am J Physiol Renal Physiol 278: F792–F798, 2000.

    Weber TJ, Liu S, Indridason OS, and Quarles LD. Serum FGF23 levels in normal and disordered phosphorus homeostasis. J Bone Miner Res 18: 1227–1234, 2003.

    Werner A, Kempson SA, Biber J, and Murer H. Increase of Na/Pi-cotransport encoding mRNA in response to low Pi diet in rat kidney cortex. J Biol Chem 269: 6637–6639, 1994.

    Werner A, Moore ML, Mantei N, Biber J, Semenza G, and Murer H. Cloning and expression of cDNA for a Na/Pi cotransport system of kidney cortex. Proc Natl Acad Sci USA 88: 9608–9612, 1991.

    White KE, Carn G, Lorenz-Depiereux B, Benet-Pages A, Strom TM, and Econs MJ. Autosomal-dominant hypophosphatemic rickets (ADHR) mutations stabilize FGF-23. Kidney Int 60: 2079–2086, 2001.

    Xiao ZS, Crenshaw M, Guo R, Nesbitt T, Drezner MK, and Quarles LD. Intrinsic mineralization defect in Hyp mouse osteoblasts. Am J Physiol Endocrinol Metab 275: E700–E708, 1998.

    Yamashita H, Yamashita T, Miyamoto M, Shigematsu T, Kazama JJ, Shimada T, Yamazaki Y, Fukumoto S, Fukagaw M, and Noguchi S. Fibroblast growth factor (FGF)-23 in patients with primary hyperparathyroidism. Eur J Endocrinol 151: 55–60, 2004.

    Yamashita T. Biological activity of FGF-23 and pathophysiologic role in chronic kidney disease. Clin Calcium 14: 760–763, 2004.

    Yamashita T, Konishi M, Miyake A, Inui K, and Itoh N. Fibroblast growth factor (FGF)-23 inhibits renal phosphate reabsorption by activation of the mitogen-activated protein kinase pathway. J Biol Chem 277: 28265–28270, 2002.

    Yamazaki Y, Okazaki R, Shibata M, Hasegawa Y, Satoh K, Tajima T, Takeuchi Y, Fujita T, Nakahara K, Yamashita T, and Fukumoto S. Increased circulatory level of biologically active full-length FGF-23 in patients with hypophosphatemic rickets/osteomalacia. J Clin Endocrinol Metab 87: 4957–4960, 2002.(Theresa J. Berndt, Susan Schiavi, and Ra)