In this issue of the Journal of Clinical Endocrinology and Metabolism, Enlund-Cerullo et al. shed some welcome light (pun intended!) on the influence of polymorphic variations in the GC gene encoding the vitamin D binding protein (DBP) on vitamin D status and the response to vitamin D supplementation in infants (1). The authors genotyped 913 healthy term infants for 4 different single nucleotide polymorphisms (SNPs) of the GC gene, and measured plasma levels of 25-hydroxyvitamin D (25(OH)D), the principal circulating form of vitamin D and the conventional biomarker for vitamin D status, in cord blood (basal state) and after daily supplementation with 10 or 30 μg of vitamin D3 from 2 weeks to 24 months of age (intervention). The authors found that minor allele homozygosity of all studied GC SNPs, their combined haplotypes and rs4588/rs7041 diplotype 2/2 were associated with lower concentrations of 25(OH)D at all time points in one or both intervention groups (analysis of covariance P < .043), with the exception of rs7041 which did not affect 25(OH)D at birth. Based on these studies, the authors conclude that in infants GC genotype affects 25(OH)D concentration and efficiency of high-dose vitamin D3 supplementation, providing further evidence that genetic background influences vitamin D status.
The present study adds to the growing evidence that nature, as well as nurture, plays an important role in vitamin D physiology. Normal vitamin D homeostasis depends upon an intricate interplay between environmental, photochemical, and biological processes. The parent forms of vitamin D, cholecalciferol (vitamin D3) and ergocalciferol (vitamin D2), may be obtained through the diet or from food supplements. Nevertheless, the chief source of vitamin D for most people is cutaneous production of cholecalciferol, which is generated by photolysis of provitamin D3 (7-dehydrocholesterol) into previtamin D3, with subsequent thermal isomerization to cholecalcifierol. This process requires exposure of the skin by UVB radiation (290–315 nm) usually from sunlight, and is diminished by increased melanin skin pigmentation and use of sunscreens or extensive body coverage (2, 3). Vitamin D is then sequentially hydroxylated in the liver and kidney by CYP2R1 (4) and CYP27B1(5), respectively, to generate 1,25-dihydroxyvitamin D (1,25(OH)2D, also known as calcitriol), the ligand for the vitamin D receptor and the metabolite that is responsible for most biological actions of vitamin D. Understanding the biology of vitamin D has led to improved ability to quantify plasma levels of vitamin D metabolites (6). On the other hand, considerable controversy persists regarding the normal level of plasma 25(OH)D. Vitamin D deficiency has historically been defined as circulating levels of 25(OH)D lower than 20 ng/mL, a reference adopted by the Institute of Medicine to meet the needs of at least 97.5% of the normal population (7). By contrast, other groups, including the Endocrine Society (8, 9), have proposed vitamin D deficiency as a 25(OH)D level that is less than 20 ng/mL with vitamin D insufficiency as 25(OH)D of 21 ng/mL to 29 ng/mL, and indicated that a 25(OH)D level of at least 30 ng/mL was required for adults to ensure bone health. These recommendations for higher levels of 25(OH)D have been influenced by a variety of human observational and association studies that have suggested a link between vitamin D deficiency and nonskeletal benefits. However, the results of randomized vitamin D intervention trials have generally been disappointing (10), thus reducing enthusiasm for additional supplementation for the general population.
Although environmental and lifestyle factors account for most of the variation in plasma levels of 25(OH)D, genetic variability also is important. Twin studies show a strong (23–80%) genetic contribution to circulating levels of 25(OH)D, but genome-wide association studies (GWAS) suggest a lesser effect of only 7 to 9%. These GWAS have identified SNPs in or near several genes involved in vitamin D homeostasis that are associated with plasma levels of 25(OH)D (11–15). Not surprisingly, these genes include CYP2R1, the principal vitamin D 25 hydroxylase, and CYP24A1, which encodes the vitamin D 24 hydroxylase that inactivates 25(OH)D. And perhaps of greatest relevance to the present work, there is also very strong association between circulating concentrations of 25(OH)D and the GC gene, which encodes the DBP. Although these SNPs have been associated with vitamin D levels, it is unknown until now if they may also be associated with the response to vitamin D supplementation. Among these genes, SNPs near GC have the greatest effect on serum vitamin 25(OH)D and therefore represent the highest possibility for targeted intervention.
GC is a highly polymorphic gene, and more than 120 GC variants have been described, although combinations of 2 common SNPs, which produce 3 major DBP isoforms (Gc1f, Gc1s, and Gc2), are present in most people and appear to segregate based on race and skin pigmentation (16). Over 90% of circulating vitamin D metabolites are tightly bound to DBP, which has a far greater apparent affinity for vitamin D metabolites than albumin, an ancestrally related protein. Therefore, it should not be surprising that conditions that result in changes in the plasma concentration of DBP (eg, pregnancy or chronic liver disease (17)) are associated with corresponding changes in circulating levels of vitamin D metabolites (17), even if levels of free 25(OH)D remain normal (18, 19). Although the authors of this study did not measure circulating levels of DBP, it has been proposed that differences in binding affinity for vitamin D metabolites may affect the plasma half-life of 25(OH)D (17, 20, 21). By contrast, different isoforms of DBP appear to have similar plasma concentrations when quantified by liquid chromatography-tandem mass spectrometry or a polyclonal enzyme-linked immunosorbent assay (22, 23).
Notwithstanding the biochemical observations reported here, it seems obvious to wonder what the biological or clinical significance might be. The free hormone hypothesis states that only unbound or free molecules are active. While free 25(OH)D may be internalized into many cells such as monocytes (20, 24), entry of 25(OH)D into proximal renal tubule cells (and parathyroid cells) is achieved by interaction of the 25(OH)D–DBP complex with the multifunctional endocytic receptor proteins megalin and cubulin (25). Once internalized, 25(OH)D3 is thought to dissociate from DBP for delivery to the renal mitochondria where it can be metabolized to 1,25(OH)2D3. The essentiality of the endocytic process for vitamin D action has been confirmed in vivo, as mice lacking megalin (26), as well as humans and dogs with loss of function mutations in cubilin (27, 28), develop signs of vitamin D deficiency secondary to impaired renal cellular uptake of 25(OH)D3–DBP and the inability to generate 1,25(OH)2D3. By contrast, complete absence of DBP in knockout mice (29) and a woman with homozygous deletion of GC genes (30) is associated with a more modest phenotype in which bone structure is relatively intact and serum levels of calcium and PTH are normal despite nearly undetectable plasma levels of 25(OH)D3 and low levels of 1,25(OH)2D. These observations might argue that the most important role that DBP serves is to buffer serum concentrations of 25(OH)D from rapid changes due to variation in vitamin D supply by prolonging the half-life of 25(OH)D in the serum.
One aspect of this work that is both a strength and weakness is the relative homogeneity of the intervention population; this decreases background genetic effects, but may also mean that this result may not be applicable to diverse populations. A second weakness is the possibility that the effects observed here only apply to high-dose supplementation of vitamin D-sufficient individuals and not to supplementation of vitamin D-deficient individuals. These issues aside, perhaps the most important aspect of this work is the refined focus on the role of GC, and other genes, on vitamin D homeostasis. In this context, genotypes for GC as well as for CYP2R1, CYP24A1, and even CYP3A4, which also can inactivate vitamin D metabolites (31, 32), may provide at least a partial explanation for the wide variation in baseline 25(OH)D levels in the normal population, as well as for the reduced response of some patients to conventional vitamin D supplementation. Notwithstanding the ongoing controversy regarding normal 25(OH)D concentrations and the wide therapeutic window for vitamin D supplementation, we submit that for at least some patients with unexplained vitamin D deficiency, an approach to vitamin D therapy that considers the patient’s specific genetic background will be advantageous. Moreover, given the growing enthusiasm for a precision medicine approach to recommendations involving diet, exercise, stress management, and dietary supplements (33), and the concept of a personal vitamin D response index (34), it is conceivable that clinicians will one day be guided by a patient’s genetic background as they calculate an optimal daily intake for vitamin D. We look forward to the next era of vitamin D biology!
Acknowledgments
Financial Support: The project described was supported by the National Institute of Diabetes and Digestive and Kidney Diseases (NIH) through NIH 5K12DK094723-03, NIH K08-HD087964-01, NIH R01DK079970. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.
Additional Information
Disclosure Summary: The authors have nothing to disclose.
Data Availability: All data generated or analyzed during this study are included in this published article or in the data repositories listed in References.
References
- 1. Enlund-Cerullo M, Koljonen L, Holmlund-Suila E, et al. Genetic variation of the vitamin D binding protein affects vitamin D status and response to supplementation in infants. J Clin Endocrinol Metab. 2019;104(11):5483–5498. [DOI] [PubMed] [Google Scholar]
- 2. Fajuyigbe D, Young AR. The impact of skin colour on human photobiological responses. Pigment Cell Melanoma Res. 2016;29(6):607–618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Passeron T, Bouillon R, Callender V, et al. Sunscreen photoprotection and vitamin D status. Br J Dermatol. 2019;181:916–931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Thacher TD, Fischer PR, Singh RJ, Roizen J, Levine MA. CYP2R1 Mutations Impair generation of 25-hydroxyvitamin D and cause an atypical form of vitamin D deficiency. J Clin Endocrinol Metab. 2015;100(7):E1005–E1013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. St-Arnaud R, Messerlian S, Moir JM, Omdahl JL, Glorieux FH. The 25-hydroxyvitamin D 1-alpha-hydroxylase gene maps to the pseudovitamin D-deficiency rickets (PDDR) disease locus. J Bone Miner Res. 1997;12(10):1552–1559. [DOI] [PubMed] [Google Scholar]
- 6. Sempos CT, Heijboer AC, Bikle DD, et al. Vitamin D assays and the definition of hypovitaminosis D: results from the First International Conference on Controversies in Vitamin D. Br J Clin Pharmacol. 2018;84(10):2194–2207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Ross AC, Manson JE, Abrams SA, et al. The 2011 report on dietary reference intakes for calcium and vitamin D from the Institute of Medicine: what clinicians need to know. J Clin Endocrinol Metab. 2011;96(1):53–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Holick MF, Binkley NC, Bischoff-Ferrari HA, et al. ; Endocrine Society . Evaluation, treatment, and prevention of vitamin D deficiency: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2011;96(7):1911–1930. [DOI] [PubMed] [Google Scholar]
- 9. Holick MF, Binkley NC, Bischoff-Ferrari HA, et al. Guidelines for preventing and treating vitamin D deficiency and insufficiency revisited. J Clin Endocrinol Metab. 2012;97(4):1153–1158. [DOI] [PubMed] [Google Scholar]
- 10. Bouillon R, Marcocci C, Carmeliet G, et al. Skeletal and extraskeletal actions of vitamin D: current evidence and outstanding questions. Endocr Rev. 2019;40(4):1109–1151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Ahn J, Yu K, Stolzenberg-Solomon R, et al. Genome-wide association study of circulating vitamin D levels. Hum Mol Genet. 2010;19(13):2739–2745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Anderson D, Holt BJ, Pennell CE, Holt PG, Hart PH, Blackwell JM. Genome-wide association study of vitamin D levels in children: replication in the Western Australian Pregnancy Cohort (Raine) study. Genes Immun. 2014;15(8):578–583. [DOI] [PubMed] [Google Scholar]
- 13. Hong J, Hatchell KE, Bradfield JP, et al. Transethnic evaluation identifies low-frequency loci associated with 25-hydroxyvitamin D concentrations. J Clin Endocrinol Metab. 2018;103(4):1380–1392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Lasky-Su J, Lange N, Brehm JM, et al. Genome-wide association analysis of circulating vitamin D levels in children with asthma. Hum Genet. 2012;131(9):1495–1505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Wang TJ, Zhang F, Richards JB, et al. Common genetic determinants of vitamin D insufficiency: a genome-wide association study. Lancet. 2010;376(9736):180–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Powe CE, Evans MK, Wenger J, et al. Vitamin D-binding protein and vitamin D status of black Americans and white Americans. N Engl J Med. 2013;369(21):1991–2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Schwartz JB, Lai J, Lizaola B, et al. A comparison of measured and calculated free 25(OH) vitamin D levels in clinical populations. J Clin Endocrinol Metab. 2014;99(5):1631–1637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Lee MJ, Kearns MD, Smith EM, Hao L, Ziegler TR, Alvarez JA, Tangpricha V. Free 25-hydroxyvitamin D concentrations in cystic fibrosis. Am J Med Sci. 2015;350(5):374–379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Schwartz JB, Gallagher JC, Jorde R, et al. Determination of free 25(OH)D concentrations and their relationships to total 25(OH)D in multiple clinical populations. J Clin Endocrinol Metab. 2018;103(9):3278–3288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Chun RF, Lauridsen AL, Suon L, et al. Vitamin D-binding protein directs monocyte responses to 25-hydroxy- and 1,25-dihydroxyvitamin D. J Clin Endocrinol Metab. 2010;95(7):3368–3376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Jones KS, Assar S, Harnpanich D, et al. 25(OH)D2 half-life is shorter than 25(OH)D3 half-life and is influenced by DBP concentration and genotype. J Clin Endocrinol Metab. 2014;99(9):3373–3381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Hoofnagle AN, Eckfeldt JH, Lutsey PL. Vitamin D-binding protein concentrations quantified by mass spectrometry. N Engl J Med. 2015;373(15):1480–1482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Denburg MR, Hoofnagle AN, Sayed S, et al. ; Chronic Renal Insufficiency Cohort study investigators . Comparison of Two ELISA methods and mass spectrometry for measurement of vitamin D-binding protein: implications for the assessment of bioavailable vitamin D concentrations across genotypes. J Bone Miner Res. 2016;31(6):1128–1136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Esteban C, Geuskens M, Ena JM, et al. Receptor-mediated uptake and processing of vitamin D-binding protein in human B-lymphoid cells. J Biol Chem. 1992;267(14):10177–10183. [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;96(4):507–515. [DOI] [PubMed] [Google Scholar]
- 26. Leheste JR, Melsen F, Wellner M, et al. Hypocalcemia and osteopathy in mice with kidney-specific megalin gene defect. Faseb J. 2003;17(2):247–249. [DOI] [PubMed] [Google Scholar]
- 27. Kook PH, Drögemüller M, Leeb T, Howard J, Ruetten M. Degenerative liver disease in young Beagles with hereditary cobalamin malabsorption because of a mutation in the cubilin gene. J Vet Intern Med. 2014;28(2):666–671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Storm T, Zeitz C, Cases O, et al. Detailed investigations of proximal tubular function in Imerslund-Gräsbeck syndrome. BMC Med Genet. 2013;14:111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Safadi FF, Thornton P, Magiera H, Hollis BW, Gentile M, Haddad JG, Liebhaber SA, Cooke NE. Osteopathy and resistance to vitamin D toxicity in mice null for vitamin D binding protein. J Clin Invest. 1999;103(2):239–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Henderson CM, Fink SL, Bassyouni H, et al. Vitamin D-binding protein deficiency and homozygous deletion of the GC gene. N Engl J Med. 2019;380(12):1150–1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Hawkes CP, Li D, Hakonarson H, Meyers KE, Thummel KE, Levine MA. CYP3A4 induction by rifampin: an alternative pathway for vitamin D inactivation in patients with CYP24A1 mutations. J Clin Endocrinol Metab. 2017;102(5):1440–1446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Roizen JD, Li D, O’Lear L, et al. CYP3A4 mutation causes vitamin D-dependent rickets type 3. J Clin Invest. 2018;128(5):1913–1918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Price ND, Magis AT, Earls JC, et al. A wellness study of 108 individuals using personal, dense, dynamic data clouds. Nat Biotechnol. 2017;35(8):747–756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Carlberg C, Haq A. The concept of the personal vitamin D response index. J Steroid Biochem Mol Biol. 2018;175:12–17. [DOI] [PubMed] [Google Scholar]