Abstract
Vitamin D metabolism consists of both production and catabolism, which are enzymatically driven and highly regulated. Renal vitamin D metabolism requires filtration and tubular reabsorption of 25-hydroxyvitamin D and is regulated by parathyroid hormone, fibroblast growth factor-23, and 1,25-dihydroxyvitamin D. In CKD, renal production of1,25-dihydroxyvitamin D from 25-hydroxyvitamin D is reduced. In addition, pharmacokinetic studies and epidemiologic studies of 24,25-dihydroxyvitamin D, the most abundant product of 25-hydroxyvitamin D catabolism by CYP24A1, suggest that vitamin D catabolism is also reduced. New insights into the mechanisms and regulation of vitamin D metabolism may lead to novel approaches to assess and treat impaired vitamin D metabolism in CKD.
Historical Context
In the early 19th century, before vitamin D was discovered, cod liver oil was used to successfully treat Rickets.1 In 1922, experiments demonstrated that the anti-rachitic substance in cod liver oil was not vitamin A, a known cod liver oil component.2 The new substance was named vitamin D because vitamins A, B, and C were already ascribed to other substances.3 In 1931, ergocalciferol (vitamin D2) was purified and crystallized. Cholecalciferol (vitamin D3) was subsequently isolated in 1935.1 It was not until the 1970s that 1,25-dihydroxyvitamin D (1,25(OH)2D) was found to be the potent hormonal form of vitamin D and that it was primarily made in the kidneys.4–8 In the 1980s, it was reported that 1,25(OH)2D was effective for the treatment of secondary hyperparathyroidism as well as osteitis fibrosa in people treated with hemodialysis.9,10 These and other seminal studies established the importance of vitamin D metabolism in bone and mineral homeostasis.
In recent years, substantial excitement and controversy have arisen regarding actions of 1,25(OH)2D on targets other than bone and mineral homeostasis.11–14 This is based on three lines of investigation. First, the vitamin D receptor has been found in most tissues of the body.11,15 Second, animal models and in vitro experiments have shown many actions of 1,25(OH)2D in tissues other than bone and the parathyroid glands.11 Third, epidemiologic research has demonstrated associations of low circulating concentrations of vitamin D metabolites (25-hydroxyvitamin D (25(OH)D) and 1,25(OH)2D) with a number of diseases, including auto-immune diseases, cardiovascular disease, and cancer.11,16–22 As a result, assessment of vitamin D-related biomarkers and treatment with vitamin D-related interventions have become common in patients with and without chronic kidney disease (CKD).19
As the clinical relevance of vitamin D has evolved, so has our understanding of vitamin D metabolism. It is now clear that multiple vitamin D metabolites are formed in the body through both activating metabolism (production) and inactivating metabolism (catabolism). This metabolism is highly regulated by an intricate endocrine system and becomes disrupted in the setting of CKD. The goal of this article is to review the current state of knowledge regarding vitamin D metabolism, focusing on new insights in its regulation and in particular on vitamin D catabolism.
Production of 1,25-dihydroxyvitamin D
Vitamin D metabolism consists of both production and catabolism, which are enzymatically driven and regulated. Vitamin D production (Figure 1) begins in the skin, where 7-dehydrocholesterol (7-DHC, a cholesterol precursor) is converted by ultraviolet radiation (UVB at a wavelength of approximately 300nm) to pre-vitamin D3. Pre-vitamin D3 is then converted by heat to vitamin D3 (cholecalciferol), which can enter the circulation, bind to vitamin D binding protein (DBP), and travel to the liver. The vitamin D3-DBP complex is taken up by the hepatocyte, where the microsomal enzyme CYP2R1 adds a hydroxyl group at the 25 position to generate 25-hydroxyvitamin D3 (25(OH)D3). 25(OH)D3 is the most abundant vitamin D metabolite in circulation, where approximately 90% is bound to DBP.23,24 In the kidney, the 25(OH)D3-DBP complex is filtered through the glomerulus into the proximal tubule and is then taken up by the proximal tubular cell via the cell surface receptors megalin and cubilin.25,26 Within the proximal tubular cell, an additional hydroxyl group may be added to 25(OH)D3 in the 1-alpha position by the mitochondrial enzyme CYP27B1, generating 1,25-dihydroxyvitamin D3 (1,25(OH)2D3).13,15 1,25(OH)D3, delivered to target tissues bound to DBP, binds to the vitamin D receptor (VDR) to regulate a wide variety of genes.
Figure 1.
Production of 1,25(OH)2D. UV-B, ultraviolet B radiation.
Vitamin D can also be taken in from the diet either as vitamin D2 (ergocalciferol) or vitamin D3 (cholecalciferol). As with skin-derived vitamin D3, diet-derived vitamin D2 and vitamin D3 are converted to 25-hydroxyvitamin D2 (25(OH)D2) or 25-hydroxyvitamin D3 (25(OH)D3), respectively, in the liver. Though subtle differences exist and may have clinical relevance, further metabolism of 25(OH)D2 generally parallels that of 25(OH)D3,.27 Therefore, throughout the remainder of this review, 25(OH)D is used to refer to 25(OH)D2 and 25(OH)D3, while 1,252(OH)D is used to refer to 1,252(OH)D2 and 1,252(OH)D3.
Catabolism of 25-hydroxyvitamin D and 1,25-dihydroxyvitamin D
Catabolism (Figure 2) is an essential and often overlooked component of vitamin D metabolism. Both 25(OH)D and 1,25(OH)2D undergo catabolism via multiple side chain hyroxylations to become more polar metabolites, which are subsequently excreted in both the urine and the feces.28 With the discovery of multiple hydroxylation products of 25(OH)D and 1,25(OH)2D, it was originally believed that vitamin D catabolism was carried out by multiple enzymes.29 Subsequent work, however, demonstrated that one enzyme, CYP24A1, is capable of catalyzing all of the hydroxylation steps in the catabolism of both 25(OH)D and 1,25(OH)2D.30–32
Figure 2.
Catabolism of 1,25(OH)2D and 25(OH)D. 25(OH)D3, 25-hydroxyvitamin D3; 1,25(OH)2D3, 1,25-dihydroxyvitamin D3; 23,25(OH)2D3, 23,25-dihydroxyvitamin D3; 24,25(OH)2D3, 24,25-dihydroxyvitamin D3; 1,23,25(OH)3D3, 1,23,25-trihydroxyvitamin D3; 1,24,25(OH)3D3, 1,24,25-trihydroxyvitamin D3; 24,25,26,27-tetranor-23(OH)D, 24,25,26,27-tetranor-23-hydroxyvitamin D.
CYP24A1 is a mitochondrial enzyme that is dependent on NADPH, adrenonexin, and adrenonexin reductase for activity.33 Structurally, it is similar to other cytochrome P450 enzymes with 12 α-helices and 4 β-sheets surrounding a heme group. It also has an aromatic cluster, which is conserved among mitochondrial P450 proteins.34 CYP24A1 catabolizes 25(OH)D to 24,25,26,27-tetranor-23-hydroxyvitamin D and 1,25(OH)2D to calcitroic acid (or via a second pathway to 23,26-lactones) through multiple hydroxylations. 30–32 This process is thought to proceed with release of the intermediate products at each step of the hydroxylation pathways, raising the possibility that measurement of intermediate products may reflect CYP24A1 activity.31
CYP24A1 has been found in many of the tissues that express the vitamin D receptor and is thought to be present in all of them.35 In the kidney, CYP24A1 is found in the proximal and distal tubule as well as the glomerular parietal epithelial cells and the glomerular mesangial cells.36 The CYP24A1 gene is highly inducible by 1,25(OH)2D in all tissues in which it is found, thus acting as a local control mechanism to prevent tissue-level 1,25(OH)2D intoxication.37 The importance of this feedback mechanism was demonstrated when inactivating mutations of CYP24A1 were identified in all eight children in a cohort with hypercalcemia after vitamin D supplementation.38 An inactivating mutation of CYP24A1 was also demonstrated in a case of an adult with hypercalcemia, elevated 1,25(OH)2D, and undetectable serum 24,25(OH)2D.39
Another P450 enzyme, CYP3A4, also plays a role in vitamin D catabolism. CYP3A4 is a microsomal cytochrome P450 found in multiple tissues including liver, small intestine, colon, kidney, esophagus, and leukocytes.40 CYP3A4 is among the most abundant cytochrome P450 enzymes in the liver, and it metabolizes the majority of drugs that undergo oxidative metabolism.41,42 In addition to drug metabolism, CYP3A4 also catabolizes 25(OH)D and 1,25(OH)2D in a manner similar to CYP24A1, with production of 1,24,25-trihydroxyvitamin D and 24,25-dihydroxyvitamin D. It also catabolizes 25(OH)D via a unique pathway, producing 4β,25-dihydroxyvitamin D.43 Interestingly, the substrate binding cavity of CYP24A1 is structurally more similar to that of CYP3A4 than any other of the known CYP protein structures.34 The quantitative contribution of CYP3A4 to vitamin D catabolism, compared with CYP24A1, is poorly defined.
Normal Regulation of vitamin D metabolism
The generation of vitamin D3 in the skin is not regulated, and bioavailability of vitamins D3 and D2 from the gastrointestinal tract is generally high.44 In addition, hepatic 25-hydroxylation of vitamins D3 and D2 is thought to be a constitutive, unregulated, process. Teleologically, regulation of these steps is probably not necessary due to the low bioactivity of these vitamin D metabolites. 25(OH)D is the first vitamin D metabolite in the activation pathway that has substantial biologic activity at the level of the vitamin D receptor, but it is at least 100-fold less active than 1,25(OH)2D.45,46
Our understanding of the regulation of 1,25(OH)2D production has increased dramatically over the past decade. Production and catabolism of 1,25(OH)2D by the kidney is tightly regulated through a complex system of hormones that together maintain calcium and phosphorus homeostasis (Figure 3). These hormones include parathyroid hormone (PTH), fibroblast growth factor-23 (FGF-23), and 1,25(OH)2D itself.
Figure 3.
Feedback regulation of vitamin D metabolism. 24,25(OH)2D3, 24,25-dihydroxyvitamin D3; 25(OH)D3, 25-hydroxyvitamin D3; 1,25(OH)2D3, 1,25-dihydroxyvitamin D3; PTH, parathyroid hormone; FGF-23, fibroblast growth factor-23.
PTH, generally secreted in response to low serum calcium, has long been known to increase circulating 1,25(OH)2D. In the kidney, PTH induces transcription of the CYP27B1 gene by stimulating protein kinase A and protein kinase C signaling pathways.47,48 PTH also down-regulates CYP24A1 mRNA transcription in the kidney by destabilizing CYP24A1 mRNA.49,50 The regulation of extra-renal 1,25(OH)2D production and catabolism by PTH is not fully elucidated, but it appears that PTH has less effect on regulation of 1,25(OH)2D production in non-renal tissues that have been examined thus far. For example, CYP27B1 expression in monocytes, respiratory epithelial cells, and epidermal keratinocytes is not regulated by PTH.15,51 Interestingly, PTH increases CYP24A1 mRNA and protein levels in transformed rat osteoblastic cells, suggesting that effects of PTH on vitamin D catabolism in bone are opposite to those in the kidney.50,52,53
FGF-23 is a recently characterized hormone produced in the bone by osteoblasts and osteocytes.54–58 It is a potent inducer of phosphaturia, which is thought to be its primary physiologic role. It also decreases CYP27B1 transcription in the kidney through the mitogen activated protein kinase (MAPK) signaling pathway and has been shown to decrease CYP27B1 renal enzyme activity.59 Furthermore, FGF-23 is a potent inducer of CYP24A1 transcription in the kidney.54,60 The effects of FGF-23 on vitamin D metabolism in extra-renal tissues are not well characterized. FGF-23 has been shown to increase CYP27B1 mRNA in cultured bovine parathyroid gland cells, which is opposite the effect of FGF-23 on CYP27B1 in the kidney.61
In the kidney, choroid plexus, and parathyroid gland, FGF-23 is thought to work only in conjunction with the membrane-bound co-factor α-klotho. Low expression of klotho has also been found in several other tissues, but its role there is uncertain. In the kidney, FGF-23 acts by binding to an FGF receptor (type 1, 3, or 4) that is coupled with α-klotho.62,63 FGF-23 also has effects on the myocardium independent of klotho.64 Furthermore, klotho is found in a soluble circulating form.65 Whether klotho has effects on regulation of vitamin D metabolism independent of FGF-23 is not known. Klotho knockout mice have increased expression of renal CYP27B1 and impaired expression of renal CYP24A1, but this is thought to be secondary to the inability of FGF-23 to act in the absence of klotho.66 Klotho does, however, have actions independent of FGF-23, as demonstrated by its ability to decrease phosphate reabsorption in the proximal tubule.65,67
In the kidney, 1,25(OH)2D is a potent regulator of its own production and catabolism through inhibition of CYP27B1 transcription and induction of CYP24A1 transcription, respectfully.68 Like PTH, 1,25(OH)2D does not appear to regulate CYP27B1 in extra-renal tissues. However, 1,25(OH)2D does potently induce CYP24A1 transcription in extra-renal tissues.15 In this way, 1,25(OH)2D tightly regulates its own intra and extra-cellular concentration to prevent 1,25(OH)2D intoxication.37
One important question that demands attention is whether vitamin D metabolites that are bound to vitamin D binding protein (DBP), bound to albumin, or unbound (free) are most clinically relevant. Experiments in mice have shown that DBP knockout mutants do not display altered calcium homeostasis or differences in target tissue concentration of 1,25(OH)2D even though they have very low total serum 25(OH)D and 1,25(OH)D concentrations.69,70 However, mice without megalin, which is necessary for renal tubular uptake of DBP, develop vitamin D deficiency and bone disease. 25 These results seem to be conflicting, with one set of experiments demonstrating that DBP is unnecessary for calcium homeostasis and the other experiments demonstrating the necessity of DBP. Recent human data offer additional insight. These studies demonstrate that the combination of free 25(OH)D and albumin-bound 25(OH)D correlate most precisely with other biomarkers of vitamin D metabolism in some populations, suggesting that this fraction of total 25(OH)D is particularly important. 71,72 Specifically, in dialysis patients, estimated free and albumin-bound 25(OH)D correlated most closely with serum corrected calcium and PTH concentrations. In young healthy adults, this fraction of 25(OH)D was more strongly associated with bone mineral density than was total 25(OH)D or free 25(OH)D alone. One potential explanation for these findings is that DBP binds 25(OH)D too tightly for it to be biologically active, while albumin binds 25(OH)D much more weakly, allowing for greater biologic availability. Furthermore, megalin mediates proximal tubular cell uptake of albumin, which could explain the findings in megalin knockout mice.73 Further research is required to determine whether measurements or estimation of specific 25(OH)D fractions is useful for clinical care.
Impaired 1,25(OH)2D production in CKD
In CKD, 1,25(OH)2D production is reduced due to alterations in CYP abundance, CYP activity, and delivery of substrate to CYP enzymes (Table 1).74–76 These mechanisms of decreased 1,25(OH)2D production are well-described though there relative contributions remain debated.
Table 1.
Effects of metabolic and hormonal alterations in CKD on vitamin D metabolism in the kidney.
| Altered CYP Abundance |
Transcription
|
Translation
|
↓CYP activity
|
↓Delivery to proximal tubular cell
|
1,25(OH)2D3, 1,25-dihydroxyvitamin D3; FGF-23, fibroblast growth factor-23; PTH, parathyroid hormone; NADPH, nicotinamide adenine dinucleotide phosphate; 25(OH)D3, 25-hydroxyvitamin D3; GFR, glomerular filtration rate.
With the recent delineation of actions of FGF-23 and the finding of markedly elevated circulating FGF-23 in CKD, there has been interest in FGF-23-mediated down-regulation of CYP27B1 as a cause of decreased 1,25(OH)2D production in CKD. Multiple studies have demonstrated that FGF-23 decreases CYP27B1 mRNA expression in the renal tubules.54,59 A rat anti-glomerular basement membrane antibody model of CKD with elevated circulating concentrations of FGF-23 and PTH demonstrated decreased renal CYP27B1 expression, increased renal CYP24A1 expression, and decreased circulating 1,25(OH)2D concentration. Administration of FGF-23 antibodies increased renal CYP27B1 mRNA, decreased renal CYP24A1 mRNA, and restored circulating 1,25(OH)2D to normal.77 These effects were not mediated by PTH, because circulating PTH concentration fell with FGF-23 antibody administration. Together, these studies consistently demonstrate that FGF-23 acts to reduce renal CYP27B1 expression. Studies have also shown that PTH increases CYP27B1 expression in renal tubular cells.47,48 Thus, two of the most markedly elevated hormones in CKD regulate CYP27B1 in a reciprocal manner. Other metabolic abnormalities commonly present in CKD, including diabetes, acidosis, and hyperuricemia, may also decrease CYP27B1 expression.78–81 Increased serum phosphorus is associated with lower circulating 1,25(OH)2D concentration, though it is not clear whether this represents a direct effect, is mediated by FGF-23, or is confounded by other factors.
Given the competing effects of FGF-23 and PTH on CYP27B1 transcription, several studies have addressed the net effects of CKD on CYP27B1 expression and activity, with mixed results. One study of 3/4 nephrectomy rats showed significantly elevated CYP27B1 mRNA expression.82 Another study of adenine-induced CKD in rats showed increased CYP27B1 mRNA and protein expression compared to normal.83 A study of human kidney biopsies demonstrated no difference in CYP27B1 expression with decreased creatinine clearance despite decreased circulating 1,25(OH)2D concentrations. This study did find, however, that increased renal inflammation was associated with elevated CYP27B1 mRNA expression in the same biopsies.84 Therefore, while FGF-23 clearly down regulates CYP27B1 transcription, it is not clear whether the decreased circulating 1,25(OH)2D concentration associated with CKD is due primarily to decreased CYP27B1 transcription induced by excess FGF-23 or related metabolic abnormalities.
Instead of, or in addition to, FGF-23-mediated reduction of CYP27B1 transcription, decreased 1,25(OH)2D production in CKD may be due to impaired delivery of 25(OH)D to CYP27B1 in the renal proximal tubule and/or decreased CYP27B1 activity. In CKD, circulating 25(OH)D concentration is often low due to obesity, low sun exposure, decreased cutaneous synthesis caused by dark skin or azotemia, and limited dietary vitamin D intake.85–87 Moreover, with declining GFR, less 25(OH)D is filtered, and therefore less 25(OH)D is available for reabsorption into the proximal tubule. In addition, absorption of filtered 25(OH)D may be impaired in CKD. Expression of megalin, which is essential for 25(OH)D uptake, is diminished in CKD,82 and with albuminuria, filtered 25(OH)D may be lost in the urine instead of undergoing proximal tubular reabsorption.88,89 Once 25(OH)D substrate is absorbed into proximal tubular cells, its metabolism to 1,25(OH)2D may still be impaired by reduced CYP27B1 activity. Diabetes (a common cause of CKD) and metabolic acidosis78 (a common result of CKD) have been associated with low circulating 1,25(OH)2D independent of circulating 25(OH)D concentration and estimated GFR.78,79 Potential explanations for these observations include diminished enzyme activity or a general reduction in proximal tubule metabolic capacity.
Impaired vitamin D catabolism in CKD
Kidney disease also disrupts vitamin D catabolism. Within the kidney, CYP24A1, like CYP27B1, is subject to competing hormonal regulation by FGF-23 and PTH. The net effects of elevated concentrations of each of these hormones on vitamin D catabolism in CKD are debated. One prevailing hypothesis posits that elevated circulating FGF-23 concentration prevails over other signals, including PTH, to induce CYP24A1 transcription, which in turn leads to accelerated 25(OH)D and 1,25(OH)2D catabolism.50,90,91 This hypothesis is based primarily on mRNA studies in rat models of CKD, which show increased CYP24A1 mRNA in diseased compared to healthy renal tissue.77,83 This mechanism has been used to explain the low circulating 25(OH)D concentrations commonly found in people with CKD. However, a study of human biopsy tissue showed no difference in CYP24A1 transcription with lower eGFR.84 Thus, neither renal CYP24A1 expression nor net renal vitamin D catabolism is completely understood in CKD.
In CKD, vitamin D catabolism may also be impaired in non-renal tissues. The relative contribution of extra-renal CYP24A1 to overall vitamin D catabolism has not been determined. It is possible that non-renal tissues contribute substantially to vitamin D catabolism, and that this contribution is decreased in CKD due to systemic 1,25(OH)2D deficiency.
Several studies have assessed the net effects of CKD on vitamin D catabolism. Horst et al measured 24,25(OH)2D in anephric and control pigs. He found that the anephric pigs were able to generate substantial circulating concentrations of 24,25(OH)2D after administration of very large doses of cholecalciferol, but that the appearance was delayed and concentrations were lower than in normal controls.92 Gray found that the half-life of 25(OH)D was twice as long in anephric versus normal humans (42 versus 23 days, respectively).28 Studies in CKD not requiring dialysis are conflicting. While Dusso observed no significant difference in the metabolic clearance rate of 1,25(OH)2D in dogs,74 Hsu found a 22% lower metabolic clearance rate of 1,25(OH)2D in humans with CKD compared to normal control subjects.93 These results suggest that extra-renal tissues are able to catabolize circulating 25(OH)D and 1,25(OH)2D, but that renal CYP24A1 is the primary site of vitamin D catabolism, which may be reduced in CKD.
24,25-dihydroxyvitamin D as a biomarker of vitamin D catabolism
Circulating 24,25(OH)2D concentration has offered new insight into vitamin D catabolism in CKD. 24,25(OH)2D is the predominant initial product of 25(OH)D catabolism by CYP24A1. Most 24,25(OH)2D generated by CYP24A1 likely quickly undergoes further metabolism by CYP24A1 to more polar metabolites. However, substantial quantities of 24,25(OH)2D appear in blood. 24,25(OH)2D generally circulates in concentrations of 1–10 ng/mL, higher than any vitamin D metabolite other than 25(OH)D and 100-fold higher than 1,25(OH)2D. In addition, 24,25(OH)2D has a circulating half-life of approximately 7 days,94 long enough that single measurements offer reasonably precise estimates of short-term blood concentrations. While the exact relationship of circulating 24,25(OH)2D concentration to CYP24A1 activity and 25(OH)D half-life has not been determined using gold standard methods, it is likely that circulating 24,25(OH)2D concentration, relative to circulating 25(OH)D concentration, provides a reasonable estimate of net 25(OH)D catabolism.
With this in mind, a number of studies have measured circulating 24,25(OH)2D concentration in order to investigate vitamin D catabolism in CKD. Several small studies in dialysis patients have shown very low to undetectable concentrations of circulating 24,25(OH)2D.28,92,95–98 Another study examined circulating 24,25(OH)2D in 76 non-dialysis CKD patients with a range of eGFR.99 This study found a direct correlation between estimated creatinine clearance and 24,25(OH)2D concentration (r = 0.25, p < 0.05). It also showed relationships of 24,25(OH)2D with 25(OH)D and serum albumin in multiple regression analysis. A limitation of these studies was measurement of 24,25(OH)2D by competitive protein binding assays. Given that differences in structures of circulating vitamin D metabolites are subtle, cross-reactivity is an important concern with antibody-based assays. For example, a recent study100 demonstrated that an antibody-based assay of plasma 1,25(OH)2D correlated poorly with plasma 1,25(OH)2D measured by mass spectrometry, suggesting that the antibody-based assay had substantial cross-reactivity with other circulating vitamin D metabolites. Therefore, we developed a novel high-throughput mass spectrometry assay for circulating 24,25(OH)2D. This assay measures 24,25(OH)2D concentration across its physiologic range (lower limit of detection 0.5ng/ml) with excellent reliability (inter-assay imprecision 8.58% at 1.5ng/ml).101
We measured serum 24,25(OH)2D concentration among 278 participants in the Seattle Kidney Study and found that lower eGFR was associated with lower circulating 24,25(OH)2D and 1,25(OH)2D concentrations but not with 25(OH)D concentration.102 Black race, diabetes, lower serum bicarbonate, lower 25(OH)D, and higher urinary albumin to creatinine ratio were also independently associated with lower 24,25(OH)2D concentration. These results suggest that CKD is a state of both decreased 1,25(OH)2D production and decreased 1,25(OH)2D catabolism.
Hypothesis: an evolving concept of impaired vitamin D metabolism in CKD
Recent insights into vitamin D regulation and catabolism suggest that CKD is a state of stagnant vitamin D metabolism characterized by reduced vitamin D catabolism and turnover in addition to reduced 1,25(OH)2D production. In this paradigm, competing effects of PTH and FGF-23 on the expression of CYP27B1 and CYP24A1 either balance each other or are superseded by a general decrease in vitamin D metabolic function in the kidney. Causes of decreased vitamin D metabolic function could include impaired uptake of 25(OH)D, diminished metabolic capacity of proximal tubular cells perhaps related to oxidative stress and redox imbalance, or a simple reduction in the number of functioning nephrons. If this hypothesis is correct, such diminished enzymatic function parallels that seen in other metabolic and signaling pathways in kidney disease including decreased drug metabolism in the gastrointestinal tract, cytochrome dysfunction in oxidative phosphorylation, dysregulated lipid metabolism, and altered growth hormone signaling.103–106 Alternatively, impaired vitamin D catabolism may be a direct consequence of reduced 1,25(OH)2D production due to reduced 1,25(OH)2D-mediated stimulation of CYP24A1. Ideally, the precise nature of impaired vitamin D metabolism and relationships of circulating hormones, other biomarkers, and treatments to vitamin D production and catabolism would be established using pharmacokinetic studies across a broad range of kidney function and race/ethnicity.
Implications for diagnostic testing
Current clinical management of bone and mineral metabolism in CKD focuses on evaluation and treatment of 25(OH)D and PTH. Recent advances make it clear that these biomarkers only partially capture vitamin D metabolism and its derangements in CKD.
Circulating 25(OH)D concentration is generally regarded as a biomarker of total vitamin D intake from cutaneous synthesis and dietary consumption, a biomarker of total body 25(OH)D stores, or both.11,19,107,108 While these interpretations are founded on high-quality studies and appear to be largely correct, they are also likely oversimplified to some extent. In particular, vitamin D catabolism may also influence steady-state blood 25(OH)D concentrations, particularly in CKD. Most studies of CKD populations report a high prevalence of 25(OH)D deficiency.99,109–113. Precise prevalence estimates vary based on geography, demographics, stage of kidney disease, and medication/supplement use. 25(OH)D deficiency is also common in the general population, and data conflict regarding whether 25(OH)D concentrations are lower in CKD than among people with normal kidney function. For example, Ravani et al found a strong positive correlation between eGFR and 25(OH)D in 168 Nephrology clinic patients with CKD.114 In contrast, in the United States population, 25(OH)D concentration was lower in people with eGFR less than 30 ml/min/1.73m2 (mean 24.7 ng/ml) but not in those with eGFR 30–59 ml/min/1.73m2 (mean 30.4 ng/ml) or eGFR 60–89 ml/min/1.73m2 (mean 31.0 ng/ml), compared with eGFR ≥90 ml/min/1.73m2 (mean 29.4 ng/ml).115 In the Study for Early Evaluation of Kidney Disease (SEEK) and the Seattle Kidney Study, 25(OH)D did not correlate with estimated GFR, while concentrations of 1,25(OH)2D, PTH, and FGF23 demonstrated strong correlations with eGFR.78,101 It is possible that conflicting associations of estimated GFR with 25(OH)D concentration relate to differences in vitamin D catabolism. In some populations, decreased 25(OH)D production due to limited ultraviolet light exposure, impaired cutaneous vitamin D synthesis,87 or dietary restriction of vitamin D-containing foods may be balanced by decreased 25(OH)D catabolism. In other populations, an imbalance of these processes may lead to changes in steady state circulating 25(OH)D concentration and/or steady state 25(OH)D body stores. Interestingly, a common genetic polymorphism in CYP24A1has been reported to influence steady-state 25(OH)D concentration in the general population.116
PTH is directly suppressed by 1,25(OH)2D.117–119 As a result, PTH is frequently used in practice as a read-out of 1,25(OH)2D activity at the level of the parathyroid gland. While using a functional measure to ascertain 1,25(OH)2D status has important strengths, this approach also has limitations. For example, PTH secretion is also controlled by other factors, including circulating calcium concentration and parathyroid gland hypertrophy. Moreover, to the extent that PTH reflects 1,25(OH)2D activity, it does so only within one organ.
Additional biomarkers are needed to better recognize, study, and treat impaired vitamin D metabolism. This may require multiple biomarkers, each measuring different aspects of the vitamin D metabolic system. A biomarker of tissue-level 1,25(OH)2D activity is particularly important but has not yet been identified. Tissue-level activity is the net result of VDR expression, 1,25(OH)2D availability, and 1,25(OH)2D catabolism. All three of these are likely to vary depending on the tissue examined. The CYP24A1 gene, which is expressed in all tissues that express VDR, is currently thought to be the most transcriptionally responsive gene to 1,25(OH)2D. To the extent that it may represent 1,25(OH)D-dependent induction of CYP24A1 in target tissues, circulating 24,25(OH)2D concentration may be a candidate biomarker for estimation of tissue-level 1,25(OH)D activity, but this has not yet been studied.
Circulating FGF-23 is also a promising biomarker for the assessment of vitamin D metabolism, and of mineral metabolism more broadly. Circulating FGF-23 concentrations increase dramatically in CKD, and higher FGF-23 concentrations are associated with increased risks of cardiovascular disease and death.120–123 In addition, FGF-23 has direct effects on the myocardium leading to hypertrophy, and therefore may be causally related to adverse outcomes.64 Currently, it remains unclear whether FGF-23 identifies individuals who respond to vitamin D-related therapies, or whether treatments that specifically target FGF-23 improve patient outcomes in CKD. Studies addressing these questions may identify new approaches to patient evaluation and treatment in the short term.
Implications for treatment
Mounting evidence suggests that disordered vitamin D metabolism has negative health consequences in CKD, but appropriate treatment strategies are not yet clear. 124 Specifically, current evidence for proper use of 1,25(OH)2D and 1,25(OH)2D analogs, nutritional vitamin D (cholecalciferol and ergocalciferol), or other novel therapies is limited.125
Active vitamin D analogs and 1,25(OH)2D have long been used to treat secondary hyperparathyroidism in patients treated with hemodialysis. More recently, animal experimental studies have demonstrated potentially beneficial effects on processes not directly related to bone and mineral homeostasis.11,14,19,51 In addition, observational studies suggest that treatment of CKD and ESRD patients with 1,25(OH)2D or active analogs reduces the risk of mortality.126–131 Randomized controlled trials to date, however, have not confirmed beneficial effects of 1,25(OH)2D or active analogs on targets beyond bone and mineral homeostasis. The VITAL study did show reduction in albuminuria and systolic blood pressure with 1–2 μg of paricalcitol daily among people with type 2 diabetes and pre-dialysis CKD, but follow-up was limited to 24 weeks.132 The PRIMO trial randomized 227 pre-dialysis CKD patients to oral paricalcitol or placebo and found no difference in left ventricular mass index during 48 weeks of follow-up.133 A Cochrane review of randomized clinical trials of 1,25(OH)2D and active analogs, which did not include data from VITAL or PRIMO, concluded that these interventions lowered PTH but had no significant effects on clinical outcomes in CKD.117
It is possible that compensatory regulatory responses to treatment with 1,25(OH)2D or active analogs reduces the benefit of this therapy. For example, circulating FGF-23 increases in response to 1,25(OH)2D administration.134 Faul et al reported that FGF-23 concentration and left ventricular hypertrophy (LVH) were independently associated in a cohort of CKD patients, and that administration of FGF-23 causes hypertrophy in isolated cardiac myocytes and LVH in klotho-deficient mice.64 In addition, vitamin D analogs induce CYP24A1, which could then lower circulating 25(OH)D and 1,25(OH)2D concentrations and mitigate some beneficial effects of treatment.
A large body of evidence now suggests that non-renal 1,25(OH)2D production is important to maintain beneficial autocrine and paracrine pathways.13,14 Therefore, circulating 25(OH)D may be an important therapeutic target. The Institute of Medicine determined that a circulating 25(OH)D concentration of 20 ng/mL was generally sufficient to maintain bone health. A recent cohort study among older adults with predominantly normal estimated GFR reported that risk of hip fracture, myocardial infarction, cancer, and death similarly increased below a threshold 25(OH)D concentration near 20 ng/mL.135 However, higher concentrations of 25(OH)D may be beneficial in CKD. In CKD, but not populations with normal estimated GFR, circulating 1,25(OH)2D and 25(OH)D concentrations directly correlate, even above 25(OH)D concentrations of 20 ng/mL, suggesting substrate dependence of CYP27B1 function above this threshold. Impaired entry of 25(OH)D into cells has been suggested by decreased megalin expression in animal models of CKD,82 and this may explain in part different thresholds by estimated GFR.
Vitamin D2 (cholecalciferol) and vitamin D3 (ergocalciferol) supplements are commonly used by people both with and without CKD and less commonly in dialysis patients. A meta-analysis reviewing the literature through November 2006 found 18 randomized controlled trials of cholecalciferol or ergocalciferol conducted predominantly among people with normal kidney function. Overall, there was decreased mortality among people taking vitamin D supplements, with a summary relative risk of death of 0.93 (95% confidence interval, 0.87 – 0.99).136 One post-hoc analysis of cholecalciferol supplementation and calcium in people with mild to moderate CKD showed improved bone mineral density at the distal radius compared to placebo.137 Other non-randomized studies of nutritional vitamin D supplementation in hemodialysis patients have shown various improvements in intermediate outcomes including increased circulating 1,25(OH)2D and albumin concentrations as well as decreased calcium, PTH, inflammatory cytokines, left ventricular mass index, and erythropoietin stimulating agent dose.138–140 Well-controlled clinical trials are needed to better test the effects of supplementation with vitamins D2 and D3 in CKD. The Vitamin D and Omega-3 Trial (VITAL), currently underway, is a large randomized 5 year trial of cancer and cardiovascular disease prevention with cholecalciferol and omega-3 fatty acids in healthy adults, selected without regards to kidney function, and it may provide further guidance on the use of vitamin D supplementation in the general population.141
Inhibition of 1,25(OH)2D catabolism has been suggested as a novel approach to vitamin D therapy in CKD.50 A CYP24A1 inhibitor has been developed, in part to mitigate the ability of cancer cells to escape the anti-proliferative effects of 1,25(OH)2D by inducing CYP24A1.142,143 This inhibitor would theoretically raise tissue and circulating 25(OH)D and 1,25(OH)2D concentrations, which could be beneficial in CKD. However, use of such an inhibitor must be approached with caution because it may exacerbate a CKD-related impairment of vitamin D catabolism and impair the ability of target tissues to protect against 1,25(OH)2D intoxication.
Future directions
While our understanding of vitamin D metabolism continues to evolve, many important aspects of diagnosis and treatment remain undefined. Physiologic studies are needed to better define vitamin D metabolism in greater detail across kidney function and other important clinical characteristics, such as race and ethnicity. Improved biomarkers are needed to diagnose perturbations in populations and individuals and to generate and test novel therapeutic interventions. Such biomarkers can also be utilized to better understand the effects of existing interventions on the intricate vitamin D metabolic system. Ultimately, this combined knowledge should be used to select promising populations, interventions, and outcomes to test whether intervening on impaired vitamin D metabolism improves clinical outcomes relevant to CKD.
Acknowledgments
Supported in part by grants R01HL096875, R01DK088762, and RC4DK090766 from the National Heart, Lung and Blood Institute and the National Institute of Diabetes and Digestive and Kidney Diseases.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Contributor Information
Cortney Bosworth, Division of Nephrology and Kidney Research Institute, University of Washington, Seattle, WA.
Ian H. de Boer, Division of Nephrology and Kidney Research Institute, Departments of Medicine and Epidemiology, University of Washington, Seattle, WA.
References
- 1.Wolf G. The discovery of vitamin D: the contribution of Adolf Windaus. J Nutr. 2004 Jun;134(6):1299–1302. doi: 10.1093/jn/134.6.1299. [DOI] [PubMed] [Google Scholar]
- 2.McCollum E, Simmonds N, Becker J, Shipley P. An experimental demonstration of the existence of a vitamin which promotes calcium deposition. The Journal of Biological Chemistry. 1922;53:293–312. [PubMed] [Google Scholar]
- 3.McCollum EV, Pitz W, Simmonds N, Becker JE, Shipley PG, Bunting RW. The effect of additions of fluorine to the diet of the rat on the quality of the teeth. 1925. Studies on experimental rickets. XXI. An experimental demonstration of the existence of a vitamin which promotes calcium deposition. 1922. The effect of additions of fluorine to the diet of the rat on the quality of the teeth. 1925. J Biol Chem. 2002 May;277(19):E8. [PubMed] [Google Scholar]
- 4.Brickman AS, Coburn JW, Norman AW. Action of 1,25-dihydroxycholecalciferol, a potent, kidney-produced metabolite of vitamin D, in uremic man. N Engl J Med. 1972 Nov;287(18):891–895. doi: 10.1056/NEJM197211022871801. [DOI] [PubMed] [Google Scholar]
- 5.Norman AW. Evidence for a new kidney-produced hormone, 1,25-dihydroxycholecalciferol, the proposed biologically active form of vitamin D. Am J Clin Nutr. 1971 Nov;24(11):1346–1351. doi: 10.1093/ajcn/24.11.1346. [DOI] [PubMed] [Google Scholar]
- 6.Omdahl J, Holick M, Suda T, Tanaka Y, DeLuca HF. Biological activity of 1,25-dihydroxycholecalciferol. Biochemistry. 1971 Jul;10(15):2935–2940. doi: 10.1021/bi00791a022. [DOI] [PubMed] [Google Scholar]
- 7.Kodicek E, Lawson DE, Wilson PW. Biological activity of a polar metabolite of vitamin D. Nature. 1970 Nov;228(5273):763–764. doi: 10.1038/228763a0. [DOI] [PubMed] [Google Scholar]
- 8.Lawson DE, Fraser DR, Kodicek E, Morris HR, Williams DH. Identification of 1,25-dihydroxycholecalciferol, a new kidney hormone controlling calcium metabolism. Nature. 1971 Mar;230(5291):228–230. doi: 10.1038/230228a0. [DOI] [PubMed] [Google Scholar]
- 9.Slatopolsky E, Weerts C, Thielan J, Horst R, Harter H, Martin KJ. Marked suppression of secondary hyperparathyroidism by intravenous administration of 1,25-dihydroxy-cholecalciferol in uremic patients. J Clin Invest. 1984 Dec;74(6):2136–2143. doi: 10.1172/JCI111639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Andress DL, Norris KC, Coburn JW, Slatopolsky EA, Sherrard DJ. Intravenous calcitriol in the treatment of refractory osteitis fibrosa of chronic renal failure. N Engl J Med. 1989 Aug;321(5):274–279. doi: 10.1056/NEJM198908033210502. [DOI] [PubMed] [Google Scholar]
- 11.Holick M. Vitamin D deficiency. N Engl J Med. 2007 Jul;357(3):266–281. doi: 10.1056/NEJMra070553. [DOI] [PubMed] [Google Scholar]
- 12.Rosen CJ, Abrams SA, Aloia JF, et al. IOM Committee Members Respond to Endocrine Society Vitamin D Guideline. J Clin Endocrinol Metab. 2012 Mar; doi: 10.1210/jc.2011-2218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Dusso A, González EA, Martin KJ. Vitamin D in chronic kidney disease. Best Pract Res Clin Endocrinol Metab. 2011 Aug;25(4):647–655. doi: 10.1016/j.beem.2011.05.005. [DOI] [PubMed] [Google Scholar]
- 14.Melamed ML, Thadhani RI. Vitamin D therapy in chronic kidney disease and end stage renal disease. Clin J Am Soc Nephrol. 2012 Feb;7(2):358–365. doi: 10.2215/CJN.04040411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Schuster I. Cytochromes P450 are essential players in the vitamin D signaling system. Biochim Biophys Acta. 2010 Jul; doi: 10.1016/j.bbapap.2010.06.022. [DOI] [PubMed] [Google Scholar]
- 16.McGreevy C, Williams D. New insights about vitamin D and cardiovascular disease: a narrative review. Ann Intern Med. 2011 Dec;155(12):820–826. doi: 10.7326/0003-4819-155-12-201112200-00004. [DOI] [PubMed] [Google Scholar]
- 17.Van Belle TL, Gysemans C, Mathieu C. Vitamin D in autoimmune, infectious and allergic diseases: a vital player? Best Pract Res Clin Endocrinol Metab. 2011 Aug;25(4):617–632. doi: 10.1016/j.beem.2011.04.009. [DOI] [PubMed] [Google Scholar]
- 18.Fleet JC, DeSmet M, Johnson R, Li Y. Vitamin D and cancer: a review of molecular mechanisms. Biochem J. 2012 Jan;441(1):61–76. doi: 10.1042/BJ20110744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Rosen CJ. Clinical practice. Vitamin D insufficiency. N Engl J Med. 2011 Jan;364(3):248–254. doi: 10.1056/NEJMcp1009570. [DOI] [PubMed] [Google Scholar]
- 20.Wang TJ, Pencina MJ, Booth SL, et al. Vitamin D deficiency and risk of cardiovascular disease. Circulation. 2008 Jan;117(4):503–511. doi: 10.1161/CIRCULATIONAHA.107.706127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Giovannucci E, Liu Y, Hollis BW, Rimm EB. 25-hydroxyvitamin D and risk of myocardial infarction in men: a prospective study. Arch Intern Med. 2008 Jun 9;168(11):1174–1180. doi: 10.1001/archinte.168.11.1174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.de Boer IH, Kestenbaum B, Shoben AB, Michos ED, Sarnak MJ, Siscovick DS. 25-hydroxyvitamin D levels inversely associate with risk for developing coronary artery calcification. J Am Soc Nephrol. 2009 Aug;20(8):1805–1812. doi: 10.1681/ASN.2008111157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Lehmann B, Meurer M. Vitamin D metabolism. Dermatol Ther. 2010 Jan;23(1):2–12. doi: 10.1111/j.1529-8019.2009.01286.x. [DOI] [PubMed] [Google Scholar]
- 24.Bikle DD, Gee E, Halloran B, Kowalski MA, Ryzen E, Haddad JG. Assessment of the free fraction of 25-hydroxyvitamin D in serum and its regulation by albumin and the vitamin D-binding protein. J Clin Endocrinol Metab. 1986 Oct;63(4):954–959. doi: 10.1210/jcem-63-4-954. [DOI] [PubMed] [Google Scholar]
- 25.Nykjaer A, Dragun D, Walther D, et al. An endocytic pathway essential for renal uptake and activation of the steroid 25-(OH) vitamin D3. Cell. 1999 Feb;96(4):507–515. doi: 10.1016/s0092-8674(00)80655-8. [DOI] [PubMed] [Google Scholar]
- 26.Nykjaer A, Fyfe JC, Kozyraki R, et al. Cubilin dysfunction causes abnormal metabolism of the steroid hormone 25(OH) vitamin D(3) Proc Natl Acad Sci U S A. 2001 Nov;98(24):13895–13900. doi: 10.1073/pnas.241516998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Bjelakovic G, Gluud LL, Nikolova D, et al. Vitamin D supplementation for prevention of mortality in adults. Cochrane Database Syst Rev. 2011;(7):CD007470. doi: 10.1002/14651858.CD007470.pub2. [DOI] [PubMed] [Google Scholar]
- 28.Gray RW, Weber HP, Dominguez JH, Lemann J. The metabolism of vitamin D3 and 25-hydroxyvitamin D3 in normal and anephric humans. J Clin Endocrinol Metab. 1974 Dec;39(6):1045–1056. doi: 10.1210/jcem-39-6-1045. [DOI] [PubMed] [Google Scholar]
- 29.Prosser DE, Jones G. Enzymes involved in the activation and inactivation of vitamin D. Trends Biochem Sci. 2004 Dec;29(12):664–673. doi: 10.1016/j.tibs.2004.10.005. [DOI] [PubMed] [Google Scholar]
- 30.Beckman MJ, Tadikonda P, Werner E, Prahl J, Yamada S, DeLuca HF. Human 25-hydroxyvitamin D3-24-hydroxylase, a multicatalytic enzyme. Biochemistry. 1996 Jun;35(25):8465–8472. doi: 10.1021/bi960658i. [DOI] [PubMed] [Google Scholar]
- 31.Sakaki T, Sawada N, Komai K, et al. Dual metabolic pathway of 25-hydroxyvitamin D3 catalyzed by human CYP24. Eur J Biochem. 2000 Oct;267(20):6158–6165. doi: 10.1046/j.1432-1327.2000.01680.x. [DOI] [PubMed] [Google Scholar]
- 32.Akiyoshi-Shibata M, Sakaki T, Ohyama Y, Noshiro M, Okuda K, Yabusaki Y. Further oxidation of hydroxycalcidiol by calcidiol 24-hydroxylase. A study with the mature enzyme expressed in Escherichia coli. Eur J Biochem. 1994 Sep;224(2):335–343. doi: 10.1111/j.1432-1033.1994.00335.x. [DOI] [PubMed] [Google Scholar]
- 33.Knutson JC, DeLuca HF. 25-Hydroxyvitamin D3-24-hydroxylase. Subcellular location and properties. Biochemistry. 1974 Mar;13(7):1543–1548. doi: 10.1021/bi00704a034. [DOI] [PubMed] [Google Scholar]
- 34.Annalora AJ, Goodin DB, Hong WX, Zhang Q, Johnson EF, Stout CD. Crystal structure of CYP24A1, a mitochondrial cytochrome P450 involved in vitamin D metabolism. J Mol Biol. 2010 Feb;396(2):441–451. doi: 10.1016/j.jmb.2009.11.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Jones G, Strugnell SA, DeLuca HF. Current understanding of the molecular actions of vitamin D. Physiol Rev. 1998 Oct;78(4):1193–1231. doi: 10.1152/physrev.1998.78.4.1193. [DOI] [PubMed] [Google Scholar]
- 36.Kumar R, Schaefer J, Grande JP, Roche PC. Immunolocalization of calcitriol receptor, 24-hydroxylase cytochrome P-450, and calbindin D28k in human kidney. Am J Physiol. 1994 Mar;266(3 Pt 2):F477–485. doi: 10.1152/ajprenal.1994.266.3.F477. [DOI] [PubMed] [Google Scholar]
- 37.Jones G. Pharmacokinetics of vitamin D toxicity. Am J Clin Nutr. 2008 Aug;88(2):582S–586S. doi: 10.1093/ajcn/88.2.582S. [DOI] [PubMed] [Google Scholar]
- 38.Schlingmann KP, Kaufmann M, Weber S, et al. Mutations in CYP24A1 and idiopathic infantile hypercalcemia. N Engl J Med. 2011 Aug;365(5):410–421. doi: 10.1056/NEJMoa1103864. [DOI] [PubMed] [Google Scholar]
- 39.Tebben PJ, Milliner DS, Horst RL, et al. Hypercalcemia, Hypercalciuria, and Elevated Calcitriol Concentrations with Autosomal Dominant Transmission Due to CYP24A1 Mutations: Effects of Ketoconazole Therapy. J Clin Endocrinol Metab. 2012 Mar;97(3):E423–427. doi: 10.1210/jc.2011-1935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Gupta RP, He YA, Patrick KS, Halpert JR, Bell NH. CYP3A4 is a vitamin D-24- and 25-hydroxylase: analysis of structure function by site-directed mutagenesis. J Clin Endocrinol Metab. 2005 Feb;90(2):1210–1219. doi: 10.1210/jc.2004-0966. [DOI] [PubMed] [Google Scholar]
- 41.Andersen MR, Farin FM, Omiecinski CJ. Quantification of multiple human cytochrome P450 mRNA molecules using competitive reverse transcriptase-PCR. DNA Cell Biol. 1998 Mar;17(3):231–238. doi: 10.1089/dna.1998.17.231. [DOI] [PubMed] [Google Scholar]
- 42.Wilkinson GR. Drug metabolism and variability among patients in drug response. N Engl J Med. 2005 May;352(21):2211–2221. doi: 10.1056/NEJMra032424. [DOI] [PubMed] [Google Scholar]
- 43.Wang Z, Lin YS, Zheng XE, et al. An Inducible Cytochrome P450 3A4-Dependent Vitamin D Catabolic Pathway. Mol Pharmacol. 2012 Apr;81(4):498–509. doi: 10.1124/mol.111.076356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Outila TA, Mattila PH, Piironen VI, Lamberg-Allardt CJ. Bioavailability of vitamin D from wild edible mushrooms (Cantharellus tubaeformis) as measured with a human bioassay. Am J Clin Nutr. 1999 Jan;69(1):95–98. doi: 10.1093/ajcn/69.1.95. [DOI] [PubMed] [Google Scholar]
- 45.Eisman JA, DeLuca HF. Intestinal 1,25-dihydroxyvitamin D3 binding protein: specificity of binding. Steroids. 1977 Aug;30(2):245–257. doi: 10.1016/0039-128x(77)90085-x. [DOI] [PubMed] [Google Scholar]
- 46.Deluca HF, Prahl JM, Plum LA. 1,25-Dihydroxyvitamin D is not responsible for toxicity caused by vitamin D or 25-hydroxyvitamin D. Arch Biochem Biophys. 2011 Jan;505(2):226–230. doi: 10.1016/j.abb.2010.10.012. [DOI] [PubMed] [Google Scholar]
- 47.Takeyama K, Kato S. The vitamin D3 1alpha-hydroxylase gene and its regulation by active vitamin D3. Biosci Biotechnol Biochem. 2011;75(2):208–213. doi: 10.1271/bbb.100684. [DOI] [PubMed] [Google Scholar]
- 48.Kim MS, Kondo T, Takada I, et al. DNA demethylation in hormone-induced transcriptional derepression. Nature. 2009 Oct;461(7266):1007–1012. doi: 10.1038/nature08456. [DOI] [PubMed] [Google Scholar]
- 49.Zierold C, Mings JA, DeLuca HF. Regulation of 25-hydroxyvitamin D3-24-hydroxylase mRNA by 1,25-ihydroxyvitamin D3 and parathyroid hormone. J Cell Biochem. 2003 Feb;88(2):234–237. doi: 10.1002/jcb.10341. [DOI] [PubMed] [Google Scholar]
- 50.Jones G, Prosser DE, Kaufmann M. 25-Hydroxyvitamin D-24-hydroxylase (CYP24A1): Its important role in the degradation of vitamin D. Arch Biochem Biophys. 2011 Nov; doi: 10.1016/j.abb.2011.11.003. [DOI] [PubMed] [Google Scholar]
- 51.Dusso AS, Tokumoto M. Defective renal maintenance of the vitamin D endocrine system impairs vitamin D renoprotection: a downward spiral in kidney disease. Kidney Int. 2011 Apr;79(7):715–729. doi: 10.1038/ki.2010.543. [DOI] [PubMed] [Google Scholar]
- 52.Huening M, Yehia G, Molina CA, Christakos S. Evidence for a regulatory role of inducible cAMP early repressor in protein kinase a-mediated enhancement of vitamin D receptor expression and modulation of hormone action. Mol Endocrinol. 2002 Sep;16(9):2052–2064. doi: 10.1210/me.2001-0260. [DOI] [PubMed] [Google Scholar]
- 53.Armbrecht HJ, Hodam TL, Boltz MA, Partridge NC, Brown AJ, Kumar VB. Induction of the vitamin D 24-hydroxylase (CYP24) by 1,25-dihydroxyvitamin D3 is regulated by parathyroid hormone in UMR106 osteoblastic cells. Endocrinology. 1998 Aug;139(8):3375–3381. doi: 10.1210/endo.139.8.6134. [DOI] [PubMed] [Google Scholar]
- 54.Shimada T, Hasegawa H, Yamazaki Y, et al. FGF-23 is a potent regulator of vitamin D metabolism and phosphate homeostasis. J Bone Miner Res. 2004 Mar;19(3):429–435. doi: 10.1359/JBMR.0301264. [DOI] [PubMed] [Google Scholar]
- 55.Yamashita T, Yoshioka M, Itoh N. Identification of a novel fibroblast growth factor, FGF-23, preferentially expressed in the ventrolateral thalamic nucleus of the brain. Biochem Biophys Res Commun. 2000 Oct;277(2):494–498. doi: 10.1006/bbrc.2000.3696. [DOI] [PubMed] [Google Scholar]
- 56.Shimada T, Mizutani S, Muto T, et al. Cloning and characterization of FGF23 as a causative factor of tumor-induced osteomalacia. Proc Natl Acad Sci U S A. 2001 May;98(11):6500–6505. doi: 10.1073/pnas.101545198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Strewler GJ. FGF23, hypophosphatemia, and rickets: has phosphatonin been found? Proc Natl Acad Sci U S A. 2001 May;98(11):5945–5946. doi: 10.1073/pnas.11154898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Tsujikawa H, Kurotaki Y, Fujimori T, Fukuda K, Nabeshima Y. Klotho, a gene related to a syndrome resembling human premature aging, functions in a negative regulatory circuit of vitamin D endocrine system. Mol Endocrinol. 2003 Dec;17(12):2393–2403. doi: 10.1210/me.2003-0048. [DOI] [PubMed] [Google Scholar]
- 59.Perwad F, Portale AA. Vitamin D metabolism in the kidney: regulation by phosphorus and fibroblast growth factor 23. Mol Cell Endocrinol. 2011 Dec;347(1–2):17–24. doi: 10.1016/j.mce.2011.08.030. [DOI] [PubMed] [Google Scholar]
- 60.Wu S, Grieff M, Brown AJ. Regulation of renal vitamin D-24-hydroxylase by phosphate: effects of hypophysectomy, growth hormone and insulin-like growth factor I. Biochem Biophys Res Commun. 1997 Apr;233(3):813–817. doi: 10.1006/bbrc.1997.6541. [DOI] [PubMed] [Google Scholar]
- 61.Krajisnik T, Björklund P, Marsell R, et al. Fibroblast growth factor-23 regulates parathyroid hormone and 1alpha-hydroxylase expression in cultured bovine parathyroid cells. J Endocrinol. 2007 Oct;195(1):125–131. doi: 10.1677/JOE-07-0267. [DOI] [PubMed] [Google Scholar]
- 62.Quarles LD. Role of FGF23 in vitamin D and phosphate metabolism: Implications in chronic kidney disease. Exp Cell Res. 2012 Mar; doi: 10.1016/j.yexcr.2012.02.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Medici D, Razzaque MS, Deluca S, et al. FGF-23-Klotho signaling stimulates proliferation and prevents vitamin D-induced apoptosis. J Cell Biol. 2008 Aug;182(3):459–465. doi: 10.1083/jcb.200803024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Faul C, Amaral AP, Oskouei B, et al. FGF23 induces left ventricular hypertrophy. J Clin Invest. 2011 Nov;121(11):4393–4408. doi: 10.1172/JCI46122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Shimamura Y, Hamada K, Inoue K, et al. Serum levels of soluble secreted α-Klotho are decreased in the early stages of chronic kidney disease, making it a probable novel biomarker for early diagnosis. Clin Exp Nephrol. 2012 Mar; doi: 10.1007/s10157-012-0621-7. [DOI] [PubMed] [Google Scholar]
- 66.Yoshida T, Fujimori T, Nabeshima Y. Mediation of unusually high concentrations of 1,25-dihydroxyvitamin D in homozygous klotho mutant mice by increased expression of renal 1alpha-hydroxylase gene. Endocrinology. 2002 Feb;143(2):683–689. doi: 10.1210/endo.143.2.8657. [DOI] [PubMed] [Google Scholar]
- 67.Hu MC, Shi M, Zhang J, et al. Klotho: a novel phosphaturic substance acting as an autocrine enzyme in the renal proximal tubule. FASEB J. 2010 Sep;24(9):3438–3450. doi: 10.1096/fj.10-154765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Akeno N, Saikatsu S, Kawane T, Horiuchi N. Mouse vitamin D-24-hydroxylase: molecular cloning, tissue distribution, and transcriptional regulation by 1alpha,25-dihydroxyvitamin D3. Endocrinology. 1997 Jun;138(6):2233–2240. doi: 10.1210/endo.138.6.5170. [DOI] [PubMed] [Google Scholar]
- 69.Safadi FF, Thornton P, Magiera H, et al. Osteopathy and resistance to vitamin D toxicity in mice null for vitamin Dbinding protein. J Clin Invest. 1999 Jan;103(2):239–251. doi: 10.1172/JCI5244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Zella LA, Shevde NK, Hollis BW, Cooke NE, Pike JW. Vitamin D-binding protein influences total circulating levels of 1,25-dihydroxyvitamin D3 but does not directly modulate the bioactive levels of the hormone in vivo. Endocrinology. 2008 Jul;149(7):3656–3667. doi: 10.1210/en.2008-0042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Bhan I, Powe CE, Berg AH, et al. Bioavailable vitamin D is more tightly linked to mineral metabolism than total vitamin D in incident hemodialysis patients. Kidney Int. 2012 Mar; doi: 10.1038/ki.2012.19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Powe CE, Ricciardi C, Berg AH, et al. Vitamin D-binding protein modifies the vitamin D-bone mineral density relationship. J Bone Miner Res. 2011 Jul;26(7):1609–1616. doi: 10.1002/jbmr.387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Cui S, Verroust PJ, Moestrup SK, Christensen EI. Megalin/gp330 mediates uptake of albumin in renal proximal tubule. Am J Physiol. 1996 Oct;271(4 Pt 2):F900–907. doi: 10.1152/ajprenal.1996.271.4.F900. [DOI] [PubMed] [Google Scholar]
- 74.Dusso A, Lopez-Hilker S, Lewis-Finch J, et al. Metabolic clearance rate and production rate of calcitriol in uremia. Kidney Int. 1989 Mar;35(3):860–864. doi: 10.1038/ki.1989.64. [DOI] [PubMed] [Google Scholar]
- 75.Gray R, Boyle I, DeLuca HF. Vitamin D metabolism: the role of kidney tissue. Science. 1971 Jun;172(989):1232–1234. doi: 10.1126/science.172.3989.1232. [DOI] [PubMed] [Google Scholar]
- 76.Fraser DR, Kodicek E. Unique biosynthesis by kidney of a biological active vitamin D metabolite. Nature. 1970 Nov;228(5273):764–766. doi: 10.1038/228764a0. [DOI] [PubMed] [Google Scholar]
- 77.Hasegawa H, Nagano N, Urakawa I, et al. Direct evidence for a causative role of FGF23 in the abnormal renal phosphate handling and vitamin D metabolism in rats with early-stage chronic kidney disease. Kidney Int. 2010 Nov;78(10):975–980. doi: 10.1038/ki.2010.313. [DOI] [PubMed] [Google Scholar]
- 78.Levin A, Bakris GL, Molitch M, et al. Prevalence of abnormal serum vitamin D, PTH, calcium, and phosphorus in patients with chronic kidney disease: results of the study to evaluate early kidney disease. Kidney Int. 2007 Jan;71(1):31–38. doi: 10.1038/sj.ki.5002009. [DOI] [PubMed] [Google Scholar]
- 79.Lee SW, Russell J, Avioli LV. 25-hydroxycholecalciferol to 1,25-dihydroxycholecalciferol: conversion impaired by systemic metabolic acidosis. Science. 1977 Mar;195(4282):994–996. doi: 10.1126/science.841324. [DOI] [PubMed] [Google Scholar]
- 80.Hsu CH, Patel SR, Young EW, Vanholder R. Effects of purine derivatives on calcitriol metabolism in rats. Am J Physiol. 1991 Apr;260(4 Pt 2):F596–601. doi: 10.1152/ajprenal.1991.260.4.F596. [DOI] [PubMed] [Google Scholar]
- 81.Vanholder R, Patel S, Hsu CH. Effect of uric acid on plasma levels of 1,25(OH)2D in renal failure. J Am Soc Nephrol. 1993 Oct;4(4):1035–1038. doi: 10.1681/ASN.V441035. [DOI] [PubMed] [Google Scholar]
- 82.Takemoto F, Shinki T, Yokoyama K, et al. Gene expression of vitamin D hydroxylase and megalin in the remnant kidney of nephrectomized rats. Kidney Int. 2003 Aug;64(2):414–420. doi: 10.1046/j.1523-1755.2003.00114.x. [DOI] [PubMed] [Google Scholar]
- 83.Helvig CF, Cuerrier D, Hosfield CM, et al. Dysregulation of renal vitamin D metabolism in the uremic rat. Kidney Int. 2010 Sep;78(5):463–472. doi: 10.1038/ki.2010.168. [DOI] [PubMed] [Google Scholar]
- 84.Zehnder D, Quinkler M, Eardley KS, et al. Reduction of the vitamin D hormonal system in kidney disease is associated with increased renal inflammation. Kidney Int. 2008 Nov;74(10):1343–1353. doi: 10.1038/ki.2008.453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Rock CL, Emond JA, Flatt SW, et al. Weight Loss Is Associated With Increased Serum 25-Hydroxyvitamin D in Overweight or Obese Women. Obesity (Silver Spring) 2012 Mar; doi: 10.1038/oby.2012.57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Bertrand KA, Giovannucci E, Liu Y, et al. Determinants of plasma 25-hydroxyvitamin D and development of prediction models in three US cohorts. Br J Nutr. 2012 Jan;:1–8. doi: 10.1017/S0007114511007409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Jacob AI, Sallman A, Santiz Z, Hollis BW. Defective photoproduction of cholecalciferol in normal and uremic humans. J Nutr. 1984 Jul;114(7):1313–1319. doi: 10.1093/jn/114.7.1313. [DOI] [PubMed] [Google Scholar]
- 88.Thrailkill KM, Jo CH, Cockrell GE, Moreau CS, Fowlkes JL. Enhanced excretion of vitamin D binding protein in type 1 diabetes: a role in vitamin D deficiency? J Clin Endocrinol Metab. 2011 Jan;96(1):142–149. doi: 10.1210/jc.2010-0980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Colston K, Williams NJ, Cleeve HJ. Studies on vitamin D binding protein in the nephrotic syndrome. Clin Chem. 1985 May;31(5):718–721. [PubMed] [Google Scholar]
- 90.Petkovich M, Jones G. CYP24A1 and kidney disease. Curr Opin Nephrol Hypertens. 2011 Jul;20(4):337–344. doi: 10.1097/MNH.0b013e3283477a7b. [DOI] [PubMed] [Google Scholar]
- 91.Quarles LD. The bone and beyond: ‘Dem bones’ are made for more than walking. Nat Med. 2011 Apr;17(4):428–430. doi: 10.1038/nm0411-428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Horst RL, Littledike ET, Gray RW, Napoli JL. Impaired 24,25-dihydroxyvitamin D production in anephric human and pig. J Clin Invest. 1981 Jan;67(1):274–280. doi: 10.1172/JCI110023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Hsu CH, Patel S, Buchsbaum BL. Calcitriol metabolism in patients with chronic renal failure. Am J Kidney Dis. 1991 Feb;17(2):185–190. doi: 10.1016/s0272-6386(12)81127-9. [DOI] [PubMed] [Google Scholar]
- 94.Leeuwenkamp O, van der Wiel H, Lips P, et al. Human pharmacokinetics of orally administered (24 R)-hydroxycalcidiol. Eur J Clin Chem Clin Biochem. 1993 Jul;31(7):419–426. doi: 10.1515/cclm.1993.31.7.419. [DOI] [PubMed] [Google Scholar]
- 95.Haddad JG, Min C, Mendelsohn M, Slatopolsky E, Hahn TJ. Competitive protein-binding radioassay of 24,25-dihydroxyvitamin D in sera from normal and anephric subjects. Arch Biochem Biophys. 1977 Aug;182(2):390–395. doi: 10.1016/0003-9861(77)90519-7. [DOI] [PubMed] [Google Scholar]
- 96.Zerwekh JE, McPhaul JJ, Parker TF, Pak CY. Extra-renal production of 24,25-dihydroxyvitamin D in chronic renal failure during 25 hydroxyvitamin D3 therapy. Kidney Int. 1983 Feb;23(2):401–406. doi: 10.1038/ki.1983.33. [DOI] [PubMed] [Google Scholar]
- 97.Weisman Y, Eisenberg Z, Leib L, Harell A, Shasha SM, Edelstein S. Serum concentrations of 24,25-dihydroxy vitamin D in different degrees of chronic renal failure. Br Med J. 1980 Sep;281(6242):712–713. doi: 10.1136/bmj.281.6242.712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Koenig KG, Lindberg JS, Zerwekh JE, Padalino PK, Cushner HM, Copley JB. Free and total 1,25-dihydroxyvitamin D levels in subjects with renal disease. Kidney Int. 1992 Jan;41(1):161–165. doi: 10.1038/ki.1992.22. [DOI] [PubMed] [Google Scholar]
- 99.Ishimura E, Nishizawa Y, Inaba M, et al. Serum levels of 1,25-dihydroxyvitamin D, 24,25-dihydroxyvitamin D, and 25-hydroxyvitamin D in nondialyzed patients with chronic renal failure. Kidney Int. 1999 Mar;55(3):1019–1027. doi: 10.1046/j.1523-1755.1999.0550031019.x. [DOI] [PubMed] [Google Scholar]
- 100.Strathmann FG, Laha TJ, Hoofnagle AN. Quantification of 1{alpha},25-Dihydroxy Vitamin D by Immunoextraction and Liquid Chromatography/Tandem Mass Spectrometry. Clin Chem. 2011 Jul; doi: 10.1373/clinchem.2010.161174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Bosworth C, Levin G, Robinson-Cohen C, et al. Serum 24,25-Dihydroxyvitamin D concentration, a Marker of Vitamin D Catabolism, is Reduced in Chronic Kidney Disease. Kidney International. 2012 doi: 10.1038/ki.2012.193. (in press) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Bosworth C, Levin G, Robinson-Cohen C, et al. Serum 24,25-Dihydroxyvitamin D concentration, a Marker of Vitamin D Catabolism, is Reduced in Chronic Kidney Disease. Kidney International. doi: 10.1038/ki.2012.193. (in press) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Granata S, Zaza G, Simone S, et al. Mitochondrial dysregulation and oxidative stress in patients with chronic kidney disease. BMC Genomics. 2009;10:388. doi: 10.1186/1471-2164-10-388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Mic J, Nolin haud TD, Naud J, et al. Effect of hemodialysis on hepatic cytochrome P450 functional expression. J Pharmacol Sci. 2008 Oct;108(2):157–163. doi: 10.1254/jphs.08042fp. [DOI] [PubMed] [Google Scholar]
- 105.Sun DF, Zheng Z, Tummala P, Oh J, Schaefer F, Rabkin R. Chronic uremia attenuates growth hormone-induced signal transduction in skeletal muscle. J Am Soc Nephrol. 2004 Oct;15(10):2630–2636. doi: 10.1097/01.ASN.0000139492.36400.6C. [DOI] [PubMed] [Google Scholar]
- 106.Hruska KA, Mathew S, Saab G. Bone morphogenetic proteins in vascular calcification. Circ Res. 2005 Jul;97(2):105–114. doi: 10.1161/01.RES.00000175571.53833.6c. [DOI] [PubMed] [Google Scholar]
- 107.Medicine Io. Dietary Reference Intakes for Calcium and Vitamin D. Washington, DC: National Acadamies Press; 2011. [PubMed] [Google Scholar]
- 108.Holick M. Vitamin D status: measurement, interpretation, and clinical application. Ann Epidemiol. 2009 Feb;19(2):73–78. doi: 10.1016/j.annepidem.2007.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.González EA, Sachdeva A, Oliver DA, Martin KJ. Vitamin D insufficiency and deficiency in chronic kidney disease. A single center observational study. Am J Nephrol 2004. 2004 Sep-Oct;24(5):503–510. doi: 10.1159/000081023. [DOI] [PubMed] [Google Scholar]
- 110.Blair D, Byham-Gray L, Lewis E, McCaffrey S. Prevalence of vitamin D [25(OH)D] deficiency and effects of supplementation with ergocalciferol (vitamin D2) in stage 5 chronic kidney disease patients. J Ren Nutr. 2008 Jul;18(4):375–382. doi: 10.1053/j.jrn.2008.04.008. [DOI] [PubMed] [Google Scholar]
- 111.LaClair RE, Hellman RN, Karp SL, et al. Prevalence of calcidiol deficiency in CKD: a cross-sectional study across latitudes in the United States. Am J Kidney Dis. 2005 Jun;45(6):1026–1033. doi: 10.1053/j.ajkd.2005.02.029. [DOI] [PubMed] [Google Scholar]
- 112.Bhan I, Burnett-Bowie SA, Ye J, Tonelli M, Thadhani R. Clinical measures identify vitamin D deficiency in dialysis. Clin J Am Soc Nephrol. 2010 Mar;5(3):460–467. doi: 10.2215/CJN.06440909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Pietrek J, Kokot F. Serum 25-hydroxyvitamin D in patients with chronic renal disease. Eur J Clin Invest. 1977 Aug;7(4):283–287. doi: 10.1111/j.1365-2362.1977.tb01606.x. [DOI] [PubMed] [Google Scholar]
- 114.Ravani P, Malberti F, Tripepi G, et al. Vitamin D levels and patient outcome in chronic kidney disease. Kidney Int. 2009 Jan;75(1):88–95. doi: 10.1038/ki.2008.501. [DOI] [PubMed] [Google Scholar]
- 115.Chonchol M, Scragg R. 25-Hydroxyvitamin D, insulin resistance, and kidney function in the Third National Health and Nutrition Examination Survey. Kidney Int. 2007 Jan;71(2):134–139. doi: 10.1038/sj.ki.5002002. [DOI] [PubMed] [Google Scholar]
- 116.Wang TJ, Zhang F, Richards JB, et al. Common genetic determinants of vitamin D insufficiency: a genome-wide association study. Lancet. 2010 Jul;376(9736):180–188. doi: 10.1016/S0140-6736(10)60588-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Palmer SC, McGregor DO, Macaskill P, Craig JC, Elder GJ, Strippoli GF. Meta-analysis: vitamin D compounds in chronic kidney disease. Ann Intern Med. 2007 Dec;147(12):840–853. doi: 10.7326/0003-4819-147-12-200712180-00004. [DOI] [PubMed] [Google Scholar]
- 118.Silver J, Naveh-Many T, Mayer H, Schmelzer HJ, Popovtzer MM. Regulation by vitamin D metabolites of parathyroid hormone gene transcription in vivo in the rat. J Clin Invest. 1986 Nov;78(5):1296–1301. doi: 10.1172/JCI112714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Russell J, Lettieri D, Sherwood LM. Suppression by 1,25(OH)2D3 of transcription of the pre-proparathyroid hormone gene. Endocrinology. 1986 Dec;119(6):2864–2866. doi: 10.1210/endo-119-6-2864. [DOI] [PubMed] [Google Scholar]
- 120.Gutierrez OM, Mannstadt M, Isakova T, et al. Fibroblast growth factor 23 and mortality among patients undergoing hemodialysis. The New England journal of medicine. 2008 Aug 7;359(6):584–592. doi: 10.1056/NEJMoa0706130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Ix JH, Shlipak MG, Wassel CL, Whooley MA. Fibroblast growth factor-23 and early decrements in kidney function: the Heart and Soul Study. Nephrol Dial Transplant. 2010 Mar;25(3):993–997. doi: 10.1093/ndt/gfp699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Kendrick J, Cheung AK, Kaufman JS, et al. FGF-23 associates with death, cardiovascular events, and initiation of chronic dialysis. J Am Soc Nephrol. 2011 Oct;22(10):1913–1922. doi: 10.1681/ASN.2010121224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Isakova T, Xie H, Yang W, et al. Fibroblast growth factor 23 and risks of mortality and end-stage renal disease in patients with chronic kidney disease. JAMA : the journal of the American Medical Association. 2011 Jun 15;305(23):2432–2439. doi: 10.1001/jama.2011.826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Slatopolsky E, Brown A, Dusso A. Pathogenesis of secondary hyperparathyroidism. Kidney Int Suppl. 1999 Dec;73:S14–19. doi: 10.1046/j.1523-1755.1999.07304.x. [DOI] [PubMed] [Google Scholar]
- 125.Group KDIGOKC-MW. KDIGO clinical practice guideline for the diagnosis, evaluation, prevention, and treatment of Chronic Kidney Disease-Mineral and Bone Disorder (CKD-MBD) Kidney Int Suppl. 2009 Aug;(113):S1–130. doi: 10.1038/ki.2009.188. [DOI] [PubMed] [Google Scholar]
- 126.Shoben AB, Rudser KD, de Boer IH, Young B, Kestenbaum B. Association of oral calcitriol with improved survival in nondialyzed CKD. J Am Soc Nephrol. 2008 Aug;19(8):1613–1619. doi: 10.1681/ASN.2007111164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Teng M, Wolf M, Ofsthun MN, et al. Activated injectable vitamin D and hemodialysis survival: a historical cohort study. J Am Soc Nephrol. 2005 Apr;16(4):1115–1125. doi: 10.1681/ASN.2004070573. [DOI] [PubMed] [Google Scholar]
- 128.Tentori F, Hunt WC, Stidley CA, et al. Mortality risk among hemodialysis patients receiving different vitamin D analogs. Kidney Int. 2006 Nov;70(10):1858–1865. doi: 10.1038/sj.ki.5001868. [DOI] [PubMed] [Google Scholar]
- 129.Shoji T, Shinohara K, Kimoto E, et al. Lower risk for cardiovascular mortality in oral 1alpha-hydroxy vitamin D3 users in a haemodialysis population. Nephrol Dial Transplant. 2004 Jan;19(1):179–184. doi: 10.1093/ndt/gfg513. [DOI] [PubMed] [Google Scholar]
- 130.Kalantar-Zadeh K, Kuwae N, Regidor DL, et al. Survival predictability of time-varying indicators of bone disease in maintenance hemodialysis patients. Kidney Int. 2006 Aug;70(4):771–780. doi: 10.1038/sj.ki.5001514. [DOI] [PubMed] [Google Scholar]
- 131.Kovesdy CP, Ahmadzadeh S, Anderson JE, Kalantar-Zadeh K. Association of activated vitamin D treatment and mortality in chronic kidney disease. Arch Intern Med. 2008 Feb;168(4):397–403. doi: 10.1001/archinternmed.2007.110. [DOI] [PubMed] [Google Scholar]
- 132.de Zeeuw D, Agarwal R, Amdahl M, et al. Selective vitamin D receptor activation with paricalcitol for reduction of albuminuria in patients with type 2 diabetes (VITAL study): a randomised controlled trial. Lancet. 2010 Nov;376(9752):1543–1551. doi: 10.1016/S0140-6736(10)61032-X. [DOI] [PubMed] [Google Scholar]
- 133.Thadhani R, Appelbaum E, Pritchett Y, et al. Vitamin D therapy and cardiac structure and function in patients with chronic kidney disease: the PRIMO randomized controlled trial. JAMA. 2012 Feb;307(7):674–684. doi: 10.1001/jama.2012.120. [DOI] [PubMed] [Google Scholar]
- 134.Liu S, Tang W, Zhou J, et al. Fibroblast growth factor 23 is a counter-regulatory phosphaturic hormone for vitamin D. J Am Soc Nephrol. 2006 May;17(5):1305–1315. doi: 10.1681/ASN.2005111185. [DOI] [PubMed] [Google Scholar]
- 135.de Boer IH, Levin G, Robinson-Cohen C, et al. Serum 25-hydroxyvitamin d concentration and risk for major clinical disease events in a community-based population of older adults: a cohort study. Annals of internal medicine. 2012 May 1;156(9):627–634. doi: 10.1059/0003-4819-156-9-201205010-00004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Autier P, Gandini S. Vitamin D supplementation and total mortality: a meta-analysis of randomized controlled trials. Arch Intern Med. 2007 Sep;167(16):1730–1737. doi: 10.1001/archinte.167.16.1730. [DOI] [PubMed] [Google Scholar]
- 137.Bosworth C, de Boer IH, Targher G, Kendrick J, Smits G, Chonchol M. The effect of combined calcium and cholecalciferol supplemenation on bone mineral density in elderly women with moderate chronic kidney disease. Clinical Nephrology. doi: 10.5414/CN107180. (in press) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Stubbs JR, Idiculla A, Slusser J, Menard R, Quarles LD. Cholecalciferol supplementation alters calcitriol-responsive monocyte proteins and decreases inflammatory cytokines in ESRD. J Am Soc Nephrol. 2010 Feb;21(2):353–361. doi: 10.1681/ASN.2009040451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Jean G, Terrat JC, Vanel T, et al. Evidence for persistent vitamin D 1-alpha-hydroxylation in hemodialysis patients: evolution of serum 1,25-dihydroxycholecalciferol after 6 months of 25-hydroxycholecalciferol treatment. Nephron Clin Pract. 2008;110(1):c58–65. doi: 10.1159/000151534. [DOI] [PubMed] [Google Scholar]
- 140.Matias PJ, Jorge C, Ferreira C, et al. Cholecalciferol supplementation in hemodialysis patients: effects on mineral metabolism, inflammation, and cardiac dimension parameters. Clin J Am Soc Nephrol. 2010 May;5(5):905–911. doi: 10.2215/CJN.06510909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Manson JE, Bassuk SS, Lee IM, et al. The VITamin D and OmegA-3 TriaL (VITAL): Rationale and design of a large randomized controlled trial of vitamin D and marine omega-3 fatty acid supplements for the primary prevention of cancer and cardiovascular disease. Contemp Clin Trials. 2011 Oct; doi: 10.1016/j.cct.2011.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Muindi JR, Yu WD, Ma Y, et al. CYP24A1 inhibition enhances the antitumor activity of calcitriol. Endocrinology. 2010 Sep;151(9):4301–4312. doi: 10.1210/en.2009-1156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Chiellini G, Rapposelli S, Zhu J, et al. Synthesis and biological activities of vitamin D-like inhibitors of CYP24 hydroxylase. Steroids. 2012 Feb;77(3):212–223. doi: 10.1016/j.steroids.2011.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]



