BookPDF Available

VITAMIN D BINDING PROTEIN, TOTAL AND FREE VITAMIN D LEVELS IN DIFFERENT PHYSIOLOGICAL AND PATHOPHYSIOLOGICAL CONDITIONS

Authors:

Figures

Content may be subject to copyright.
EDITED BY : Daniel David Bikle, Zhongjian Xie and Xiangbing Wang
PUBLISHED IN : Frontiers in Endocrinology
VITAMIN D BINDING PROTEIN, TOTAL AND
FREE VITAMIN D LEVELS IN DIFFERENT
PHYSIOLOGICAL AND PATHOPHYSIOLOGICAL
CONDITIONS
Frontiers in Endocrinology
1
March 2020 | Vitamin D Binding Protein
About Frontiers
Frontiers is more than just an open-access publisher of scholarly articles: it is a
pioneering approach to the world of academia, radically improving the way scholarly
research is managed. The grand vision of Frontiers is a world where all people have
an equal opportunity to seek, share and generate knowledge. Frontiers provides
immediate and permanent online open access to all its publications, but this alone
is not enough to realize our grand goals.
Frontiers Journal Series
The Frontiers Journal Series is a multi-tier and interdisciplinary set of open-access,
online journals, promising a paradigm shift from the current review, selection and
dissemination processes in academic publishing. All Frontiers journals are driven
by researchers for researchers; therefore, they constitute a service to the scholarly
community. At the same time, the Frontiers Journal Series operates on a revolutionary
invention, the tiered publishing system, initially addressing specific communities of
scholars, and gradually climbing up to broader public understanding, thus serving
the interests of the lay society, too.
Dedication to Quality
Each Frontiers article is a landmark of the highest quality, thanks to genuinely
collaborative interactions between authors and review editors, who include some
of the world’s best academicians. Research must be certified by peers before entering
a stream of knowledge that may eventually reach the public - and shape society;
therefore, Frontiers only applies the most rigorous and unbiased reviews.
Frontiers revolutionizes research publishing by freely delivering the most outstanding
research, evaluated with no bias from both the academic and social point of view.
By applying the most advanced information technologies, Frontiers is catapulting
scholarly publishing into a new generation.
What are Frontiers Research Topics?
Frontiers Research Topics are very popular trademarks of the Frontiers Journals
Series: they are collections of at least ten articles, all centered on a particular subject.
With their unique mix of varied contributions from Original Research to Review
Articles, Frontiers Research Topics unify the most influential researchers, the latest
key findings and historical advances in a hot research area! Find out more on how
to host your own Frontiers Research Topic or contribute to one as an author by
contacting the Frontiers Editorial Oce: researchtopics@frontiersin.org
Frontiers eBook Copyright Statement
The copyright in the text of
individual articles in this eBook is the
property of their respective authors
or their respective institutions or
funders. The copyright in graphics
and images within each article may
be subject to copyright of other
parties. In both cases this is subject
to a license granted to Frontiers.
The compilation of articles
constituting this eBook is the
property of Frontiers.
Each article within this eBook, and
the eBook itself, are published under
the most recent version of the
Creative Commons CC-BY licence.
The version current at the date of
publication of this eBook is
CC-BY 4.0. If the CC-BY licence is
updated, the licence granted by
Frontiers is automatically updated to
the new version.
When exercising any right under the
CC-BY licence, Frontiers must be
attributed as the original publisher
of the article or eBook, as
applicable.
Authors have the responsibility of
ensuring that any graphics or other
materials which are the property of
others may be included in the
CC-BY licence, but this should be
checked before relying on the
CC-BY licence to reproduce those
materials. Any copyright notices
relating to those materials must be
complied with.
Copyright and source
acknowledgement notices may not
be removed and must be displayed
in any copy, derivative work or
partial copy which includes the
elements in question.
All copyright, and all rights therein,
are protected by national and
international copyright laws. The
above represents a summary only.
For further information please read
Frontiers’ Conditions for Website
Use and Copyright Statement, and
the applicable CC-BY licence.
ISSN 1664-8714
ISBN 978-2-88963-578-8
DOI 10.3389/978-2-88963-578-8
Frontiers in Endocrinology
2
March 2020 | Vitamin D Binding Protein
VITAMIN D BINDING PROTEIN, TOTAL AND
FREE VITAMIN D LEVELS IN DIFFERENT
PHYSIOLOGICAL AND PATHOPHYSIOLOGICAL
CONDITIONS
Topic Editors:
Daniel David Bikle, University of California, San Francisco, United States
Zhongjian Xie, Central South University, China
Xiangbing Wang, Rutgers, The State University of New Jersey, United States
Citation: Bikle, D. D., Xie, Z., Wang, X., eds. (2020). Vitamin D Binding Protein,
Total and Free Vitamin D Levels in Dierent Physiological and Pathophysiological
Conditions. Lausanne: Frontiers Media SA. doi: 10.3389/978-2-88963-578-8
Frontiers in Endocrinology
3
March 2020 | Vitamin D Binding Protein
04 Editorial: Vitamin D Binding Protein, Total and Free Vitamin D Levels in
Different Physiological and Pathophysiological Conditions
Zhongjian Xie, Xiangbing Wang and Daniel D. Bikle
07 Association of Vitamin D Metabolites With Embryo Development and
Fertilization in Women With and Without PCOS Undergoing Subfertility
Treatment
Thomas Keith Cunningham, Victoria Allgar, Soha R. Dargham, Eric Kilpatrick,
Thozhukat Sathyapalan, Stephen Maguiness, Haira R. Mokhtar Rudin,
Nour M. Abdul Ghani, Aishah Latiff and Stephen L. Atkin
14 The Role of Vitamin D Binding Protein, Total and Free 25-Hydroxyvitamin
D in Diabetes
Rolf Jorde
21 25-Hydroxyvitamin D and Vitamin D Binding Protein Levels in Patients
With Primary Hyperparathyroidism Before and After Parathyroidectomy
Xiangbing Wang, Zhifeng Sheng, Lingqiong Meng, Chi Su, Stanley Trooskin
and Sue A. Shapses
27 Vitamin D Binding Protein, Total and Free Vitamin D Levels in Different
Physiological and Pathophysiological Conditions
Daniel David Bikle and Janice Schwartz
39 The Vitamin D Binding Protein and Inflammatory Injury: A Mediator or
Sentinel of Tissue Damage?
Richard R. Kew
49 Vitamin D Binding Protein and the Biological Activity of Vitamin D
Rene F. Chun, Albert Shieh, Carter Gottlieb, Vahe Yacoubian, Jeffrey Wang,
Martin Hewison and John S. Adams
64 Vitamin D Binding Protein: A Historic Overview
Roger Bouillon, Frans Schuit, Leen Antonio and Fraydoon Rastinejad
Table of Contents
EDITORIAL
published: 11 February 2020
doi: 10.3389/fendo.2020.00040
Frontiers in Endocrinology | www.frontiersin.org 1February 2020 | Volume 11 | Article 40
Edited and reviewed by:
Jonathan H. Tobias,
University of Bristol, United Kingdom
*Correspondence:
Zhongjian Xie
zhongjian.xie@csu.edu.cn
Specialty section:
This article was submitted to
Bone Research,
a section of the journal
Frontiers in Endocrinology
Received: 13 January 2020
Accepted: 22 January 2020
Published: 11 February 2020
Citation:
Xie Z, Wang X and Bikle DD (2020)
Editorial: Vitamin D Binding Protein,
Total and Free Vitamin D Levels in
Different Physiological and
Pathophysiological Conditions.
Front. Endocrinol. 11:40.
doi: 10.3389/fendo.2020.00040
Editorial: Vitamin D Binding Protein,
Total and Free Vitamin D Levels in
Different Physiological and
Pathophysiological Conditions
Zhongjian Xie 1
*, Xiangbing Wang 2and Daniel D. Bikle3
1Hunan Provincial Key Laboratory of Metabolic Bone Diseases, National Clinical Research Center for Metabolic Diseases,
Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China,
2Division of Endocrinology, Metabolism and Nutrition, Rutgers University-Robert Wood Johnson Medical School,
New Brunswick, NJ, United States, 3Endocrine Unit, Veterans Affairs Medical Center, University of California, San Francisco,
San Francisco, CA, United States
Keywords: vitamin D binding protein, total 25-hydroxyvitamin D, free 25-hydroxyvitamin D, 1,25-dihydroxyvitamin
D, bioavailable 25-hydroxyvitamin D
Editorial on the Research Topic
Vitamin D Binding Protein, Total and Free Vitamin D Levels in Different Physiological and
Pathophysiological Conditions
Vitamin D binding protein (DBP) is a major plasma carrier for vitamin D and its metabolites.
In recent years, there has been growing interest in understanding the physiological functions and
attributes of DBP. The current issue is comprised of five review articles and two original research
papers concerning the physiology of DBP and its role in different disorders.
Poor vitamin D status is highly prevalent in many different countries (14), but the
exact definition of vitamin D status is controversial. The plasma concentration of total
25-hydroxyvitamin D [25(OH)D] is currently used as an indicator of vitamin D status. In the
past decades, however, there has been argument as to whether just measuring total 25(OH)D is
appropriate for the assessment of vitamin D status in different physiological and pathophysiological
conditions (5,6). About 85% of the total circulating 25(OH)D is bound to DBP, and 15% is
bound to albumin. About 0.03% of 25(OH)D circulates in free form. Since 25(OH)D is weakly
bound to albumin and dissociates from it during tissue perfusion, the sum of the free and the
albumin-bound 25(OH)D represents the bioavailable 25(OH)D, which may be readily available for
metabolic function. In contrast, the DBP-bound vitamin D is relatively unavailable to target tissue,
with the exception of a few tissues such as the kidney that express a megalin/cubilin transport
system for DBP-bound 25(OH)D. The concept that it is the free hormone and not the DBP-
bound hormone that enters cells is known as the free hormone hypothesis. In the review by Bikle
and Schwartz it is highlighted that the DBP level is regulated by estrogen, glucocorticoids, and
inflammatory cytokines but not by vitamin D itself, and therefore, these regulators would affect
levels of total 25(OH)D. The review by Bikle and Schwartz focuses on the biological importance
of DBP with emphasis on its regulation of total and free vitamin D metabolite levels in various
clinical conditions. They also point out that attempts to calculate the free level using affinity
constants generated in a normal individual along with measurement of DBP and total 25(OH)D
have not accurately reflected directly measured free levels in a number of clinical conditions. The
authors examine the impact of different clinical conditions as well as different DBP alleles on
the relationship between total and free 25(OH)D, using only data in which the free 25(OH)D
4
Xie et al. Vitamin D Binding Protein
level was directly measured. Following their previous review (7),
the review by Chun et al. discussed a number of important
questions including the following. Is the total 25(OH)D (bound
plus free) or the unbound free 25(OH)D the crucial determinant
of the non-classical actions of vitamin D? While DBP-bound
25(OH)D is important for renal handling of 25(OH)D and
endocrine synthesis of 1,25(OH)2D, how does DBP impact extra-
renal synthesis of 1,25(OH)2D and subsequent 1,25(OH)2D
actions? Are there pathophysiological contexts where total
25(OH)D and free 25(OH)D would diverge in value as a marker
of vitamin D status? This review aims to introduce the concept
of free 25(OH)D and the molecular biology and biochemistry
of vitamin D and DBP, which provides the context for free
25(OH)D, and surveys in vitro, animal, and human studies taking
free 25(OH)D into consideration.
Low DBP levels in patients with primary hyperparathyroidism
(PHPT) were first reported in 2013 by Wang’s group (8) and
confirmed by Battista et al. (9). In the paper by Wang et al.,
the authors recruited 75 patients with PHPT and 75 healthy
control subjects. In addition, 25 PHPT patients underwent
parathyroidectomy and had a 3-month follow up visit. The
results showed that serum DBP levels were lower in patients with
PHPT but that parathyroidectomy restored DBP levels. Lower
DBP levels may be one of the contributing factors of low total
25(OH)D level in PHPT patients, and the total 25(OH)D levels
might not reflect true vitamin D status in patients with PHPT.
In the comprehensive review by Bouillon et al. it was noted
that DBP was originally discovered as a highly polymorphic
protein useful for population studies and originally called Group-
specific Component (GC). It is now known that DBP and
GC are the same protein and appeared early in the evolution
of vertebrates. DBP is genetically the oldest member of the
albuminoid family (which includes albumin, α-fetoprotein, and
afamin, all involved in the transport of fatty acids or hormones).
DBP has a single binding site for all vitamin D metabolites
with a high affinity for 25(OH)D, thereby creating a large
pool of circulating 25(OH)D, which prevents rapid vitamin D
deficiency. The review also highlighted the roles of DBP in
preventing the urinary loss of 25(OH)D and the formation of
polymeric actin fibrils in the circulation after tissue damage.
DBP also plays a minor role in transporting fatty acids. Based
on the fact that the total concentrations of 25(OH)D and
1,25(OH)2D in DBP null mice or humans are extremely low but
calcium and bone homeostasis remain normal, the “free hormone
hypothesis” appears to apply to the vitamin D hormones,
25(OH)D or 1,25(OH)2D, as it does to other steroid hormones
and thyroid hormone.
Vitamin D is important for bone health but may also
have extra-skeletal effects. Cunningham et al. examined vitamin
Dmetabolites in serum samples from age- and weight-
matched women with and without PCOS and reported results
in their paper. The authors found that 25-hydroxy-3epi-
Vitamin D3, 25-hydroxyvitamin D2, 25-hydroxyvitamin D3, and
24,25-dihydroxyvitamin D3, but not 1,25-dihydroxyvitamin D3
[1,25(OH)2D3], were associated with embryo parameters. The
data suggest that vitamin D metabolites other than 1,25(OH)2D3
are important in fertility. Kew, in his review assesses the
fundamental role of DBP in neutrophilic inflammation and
injury. As highlighted by Kew, DBP induces selective recruitment
of neutrophils. DBP is also an extracellular scavenger for actin
released from damaged/dead cells, and formation of DBP-actin
complexes is an immediate host response to tissue injury. DBP
bound to G-actin functions as an indirect but essential cofactor
for neutrophil migration.
Vitamin D and DBP have immunological effects and may
be important in the development of type 1 diabetes (T1DM).
Moreover, low total 25(OH)D levels are associated with the
development of type 2 diabetes (T2DM). However, there are no
convincing data showing that vitamin D supplementation has
an effect on the prevention of T2DM (10). The review by Jorde
discusses the relations between DBP and total and free 25(OH)D
in T1DM and T2DM.
AUTHOR CONTRIBUTIONS
All authors listed have made a substantial, direct and intellectual
contribution to the work, and approved it for publication.
FUNDING
This study was supported by National Natural Science
Foundation of China (81672646, 81471055).
REFERENCES
1. Xie Z, Xia W, Zhang Z, Wu W, Lu C, Tao S, et al. Prevalence
of vitamin D inadequacy among chinese postmenopausal women: a
nationwide, multicenter, cross-sectional study. Front Endocrinol. (2007) 9:782.
doi: 10.3389/fendo.2018.00782
2. Hypponen E, Power C. Hypovitaminosis D in British adults at age 45 y:
nationwide cohort study of dietary and lifestyle predictors. Am J Clin Nutr.
(2007) 85:860–8. doi: 10.1093/ajcn/85.3.860
3. Byun EJ, Heo J, Cho S, Lee HJD, Kim HS. Suboptimal vitamin D
status in Korean adolescents: a nationwide study on its prevalence,
risk factors including cotinine-verified smoking status and association
with atopic dermatitis and asthma. BMJ Open. (2017) 7:e016409.
doi: 10.1136/bmjopen-2017-016409
4. Acherjya GK, Ali M, Tarafder K, Akhter N, Chowdhury MK, Islam DU, et al.
Study of vitamin D deficiency among the apparently healthy population in
Jashore, Bangladesh. Mymensingh Med J. (2019) 28:214–21.
5. Youselzadeh P, Shapese SA, Wang X. Vitamin D binding protein impact
on 25-hydroxyvitamin D levels under different physiologic and pathologic
conditions. Int J Endocrinol. (2014) 2014:981581. doi: 10.1155/2014/
981581
6. Jassil N, Sharma A, Bikle DD, Wang X. Vitamin D binding protein and 25-
hydroxyvitamin D levels: emerging clinical applications. Endocr Pract. (2017)
23:605–61. doi: 10.4158/EP161604.RA
7. Chun RF, Peercy BE, Orwoll ES, Nielson CM, Adams JS, Hewison M.
Vitamin D and DBP: the free hormone hypothesis revisited. J Steroid
Biochem Mol Biol. (2014) 144 Pt A:132–7. doi: 10.1016/j.jsbmb.2013.
09.012
Frontiers in Endocrinology | www.frontiersin.org 2February 2020 | Volume 11 | Article 40
5
Xie et al. Vitamin D Binding Protein
8. Wang X, Shapses SA, Wei S, Sukumar D, Ghosh J. Vitamin D-binding protein
levels in female patients with primary hyperparathyroidism. Endocr Pract.
(2013) 19:609–13. doi: 10.4158/EP12371.OR
9. Battista C, Guarnieri V, Carnevale V, Baorda F, Pileri M, Garrubba M,
et al. Vitamin D status in primary hyperparathyroidism: effect of genetic
background. Endocrine. (2017) 55:266–72. doi: 10.1007/s12020-016-0974-x
10. Pittas AG, Dawson-Hughes B, Sheehan P, Ware JH, Knowler WC,
Aroda VR, et al. Vitamin D Supplementation and Prevention of Type
2 Diabetes. N Engl J Med. (2019) 381:520–30. doi: 10.1056/NEJMoa19
00906
Conflict of Interest: The authors declare that the research was conducted in the
absence of any commercial or financial relationships that could be construed as a
potential conflict of interest.
Copyright © 2020 Xie, Wang and Bikle. This is an open-access article distributed
under the terms of the Creative Commons Attribution License (CC BY). The use,
distribution or reproduction in other forums is permitted, provided the original
author(s) and the copyright owner(s) are credited and that the original publication
in this journal is cited, in accordance with accepted academic practice. No use,
distribution or reproduction is permitted which does not comply with these terms.
Frontiers in Endocrinology | www.frontiersin.org 3February 2020 | Volume 11 | Article 40
6
ORIGINAL RESEARCH
published: 29 January 2019
doi: 10.3389/fendo.2019.00013
Frontiers in Endocrinology | www.frontiersin.org 1January 2019 | Volume 10 | Article 13
Edited by:
Daniel David Bikle,
University of California, San Francisco,
United States
Reviewed by:
Carol L. Wagner,
Medical University of South Carolina,
United States
Michael F. Holick,
Boston Medical Center, United States
*Correspondence:
Stephen L. Atkin
sla2002@qatar-med.cornell.edu
Specialty section:
This article was submitted to
Bone Research,
a section of the journal
Frontiers in Endocrinology
Received: 02 October 2018
Accepted: 10 January 2019
Published: 29 January 2019
Citation:
Cunningham TK, Allgar V,
Dargham SR, Kilpatrick E,
Sathyapalan T, Maguiness S, Mokhtar
Rudin HR, Abdul Ghani NM, Latiff A
and Atkin SL (2019) Association of
Vitamin D Metabolites With Embryo
Development and Fertilization in
Women With and Without PCOS
Undergoing Subfertility Treatment.
Front. Endocrinol. 10:13.
doi: 10.3389/fendo.2019.00013
Association of Vitamin D Metabolites
With Embryo Development and
Fertilization in Women With and
Without PCOS Undergoing
Subfertility Treatment
Thomas Keith Cunningham 1,2 , Victoria Allgar 3, Soha R. Dargham 4, Eric Kilpatrick 5,
Thozhukat Sathyapalan 2, Stephen Maguiness 1, Haira R. Mokhtar Rudin 6,
Nour M. Abdul Ghani 6, Aishah Latiff 6and Stephen L. Atkin 4
*
1Hull IVF Unit, Women and Children’s Hospital, Hull Royal Infirmary, Hull, United Kingdom, 2Centre for Diabetes and
Metabolic Research, Hull York Medical School, University of Hull, Hull, United Kingdom, 3Department of Statistics, Hull York
Medical School, University of Hull, Hull, United Kingdom, 4Weill Cornell Medicine Qatar, Doha, Qatar, 5Sidra Medical and
Research Centre, Doha, Qatar, 6Antidoping Laboratory Qatar, Doha, Qatar
Objective: The relationship between fertilization rates and 1,25-dihydroxyvitamin D
(1,25(OH)2D3), 25-hydroxyvitamin D2 (25(OH)D2), 25-hydroxyvitamin D3 (25(OH)D3),
24,25-dihydroxyvitamin D (24,25(OH)2D3), and 25-hydroxy-3epi-Vitamin D3
(3epi25(OH)D3) concentrations in age and weight matched women with and without
PCOS was studied.
Methods: Fifty nine non-obese women, 29 with PCOS, and 30 non-PCOS undergoing
IVF, matched for age and weight were included. Serum vitamin D metabolites were taken
the menstrual cycle prior to commencing controlled ovarian hyperstimulation.
Results: Vitamin D metabolites did not differ between PCOS and controls; however,
25(OH)D3correlated with embryo fertilization rates in PCOS patients alone (p=0.03).
For all subjects, 3epi25(OH)D3correlated with fertilization rate (p<0.04) and negatively
with HOMA-IR (p<0.02); 25(OH)D2correlated with cleavage rate, G3D3 and blastocyst
(p<0.05; p<0.009; p<0.002, respectively). 24,25(OH)2D3correlated with AMH,
antral follicle count, eggs retrieved and top quality embryos (G3D3) (p<0.03; p<
0.003; p<0.009; p<0.002, respectively), and negatively with HOMA-IR (p<0.01).
1,25(OH)2D3did not correlate with any of the metabolic or embryo parameters. In slim
PCOS, 25(OH)D3correlated with increased fertilization rates in PCOS, but other vitamin
D parameters did not differ to matched controls.
Conclusion: 3epi25(OH)D3, 25(OH)D2, and 24,25(OH)2D3, but not 1,25(OH)2D3, were
associated with embryo parameters suggesting that vitamin D metabolites other than
1,25(OH)2D3are important in fertility.
Keywords: vitamin D, vitamin D epimers, vitamin D metabolites, fertilization rates, PCOS
7
Cunningham et al. Vitamin D and Embryo Development in PCOS
INTRODUCTION
Polycystic ovarian syndrome (PCOS) is one of the most
common endocrine disorders amongst women of reproductive
age affecting 9–21% of the female population and is the main
cause of anovulatory infertility (1). It is associated with clinical
and biochemical hyperandrogenism, and insulin resistance (IR)
in PCOS is associated with obesity, type 2 diabetes, and
hypercholestrolemia (2). Vitamin D levels are low in 67–85% of
women with PCOS (3), which are suggested to exacerbate IR
and the free androgen index (FAI) in PCOS (4,5). IR itself is
both independent of and exacerbated by obesity and is present
in 65–80% of women with PCOS (6) and may be improved by
vitamin D replacement (7). In a recent meta-analysis, it was
shown that in weight matched PCOS women, vitamin D was
negatively predicted by weight hip ratio, glucose and LH (8).
Vitamin D deficiency has become the most common
nutritional deficiency throughout the world (9). Studies of sub-
fertile women have demonstrated that vitamin D deficiency
is present in between 58 and 91% of cases (912). Obesity
can exacerbate vitamin D deficiency, as a result of decreased
bioavailability from cutaneous and dietary sources because of
deposition in the body fat compartments (13).
Vitamin D3 (cholecalciferol) is endogenously produced or
taken as a dietary supplement, whist vitamin D2 (ergocalciferol)
is derived from the diet (primarily from mushrooms and fungi),
though both are hydroxylated to 25(OH)D3or 25(OH)D2
by multiple 25-hydroxylases (14,15) (Figure 1). 25(OH)D is
transported to the kidney and converted to either the active 1,25-
dihydroxyvitamin D (1,25(OH)2D3) by 1 alpha hydroxylase, or
to 24,25-dihydroxyvitamin D (24,25(OH)2D3), that is also active,
by the 24 alpha hydroxylase, in the renal tubular and other
cells widely in the body (Figure 1) (16). It has been recently
reported that extrarenal tissues may also convert 25(OH)D
to 1,25(OH)2D (17). 1,25(OH)2D binds to the vitamin D
receptor (VDR) subsequently heterodimerizes with the retinoid
X receptor for its action that may be effected in several hours (16);
however, a more rapid action has been reported with binding
membrane VDR or through the 1,25D3-membrane-associated,
rapid response steroid-binding protein receptor with activation
of protein kinases A and C (18). Vitamin D receptors have
been located within structures of the female reproductive tract,
including the ovary and endometrium (19,20).
Vitamin D2is derived from the diet as ergocalciferol
that has lower binding efficacy to VDR resulting in greater
serum clearance, limiting the formation of 25(OH)D2, though
1,25(OH)2D2has a high an affinity for VDR as 1,25(OH)2D3
(14,16). In the United States and other countries vitamin D2 is
available both as a supplement and as a pharmaceutical to treat
vitamin D deficiency.
3- epimerase isomerizes the C-3 hydroxy group of the
natural vitamin D from the αto the βorientation leading to
3epi25(OH)D3(14,21) that may be measured inadvertently
whilst measuring 25(OH)D3(22). 3epi25(OH)D3is thought to
be less potent physiologically as 25(OH)D3, and 1,25(OH)2-3-
epi-D3has less affinity to VDR thus less biologically active;
however, the 3-epimer may be as potent as 1,25(OH)2D3in other
circumstances such as PTH suppression (14,23) however, data is
sparse on the biological potency of the C3 epimers.
Two IVF cohort studies have suggested that clinical pregnancy
rates were significantly lower in women who were vitamin
D deficient (24,25), but no differences in the embryological
data have been associated with 25(OH)D3levels (25). However,
it remains unknown if there is a relationship of baseline
1,25(0H)2D3, 25(OH)D3, 25(OH)D2, 24,25(0H)2D3, or its
epimer 3epi25(OH)D3to fertilization in non-obese PCOS
subjects when age and weight are matched to control subjects,
and therefore this study was undertaken.
MATERIALS AND METHODS
This prospective cohort study was performed within the Hull IVF
Unit, UK following approval by the Yorkshire and The Humber
NRES ethical committee, UK and all gave their written informed
consent. The PCOS subjects were recruited using the revised 2003
criteria (26), namely any 2 out of 3 criteria were met; menstrual
disturbance (oligo or amenorrhoea), clinical and/or biochemical
signs of androgenism and polycystic ovaries on ultrasound, with
the exclusion of other conditions. All women were on folic
acid 400 mcg daily but no other medication. Exclusion criteria
were patients with diabetes, renal or liver insufficiency, acute or
chronic infections, systemic inflammatory diseases, age <20, age
>45, known Immunological disease.
Sample Collection
A fasting blood sample was taken in the luteal phase of the
cycle before commencing IVF treatment. The bloods were
centrifuged at 3,500 g for 15 min and placed into aliquots and
frozen at 80C until analysis. The bloods were analyzed
for FSH (Architect analyser, Abbott laboratories, Maidenhead,
United Kingdom), SHBG, insulin (DPC Immulite 200 analyser,
Euro/DPC, Llanberis United Kingdom), and plasma glucose
(Synchron LX20 analyser, Beckman-Coulter, High Wycombe,
United Kingdom). Free androgen index (FAI) was calculated by
dividing the total testosterone by SHBG, and then multiplying
by one hundred. Insulin resistance (IR) was calculated using
the homeostasis model assessment (HOMA-IR). Serum vitamin
D levels and testosterone were quantified using isotope-
dilution liquid chromatography tandem mass spectrometry (LC-
MS/MS). Vitamin D metabolites (1,25(OH)2D3), 25(OH)D2,
25(OH)D3, 24,25(OH)2D3 and 3epi25(OH)D3 and three labeled
internal standards (d6-25(OH)D3, d6-1,25(OH)2D3 and d6-3-
epi-25(OH)D3) were simultaneously extracted from 250 µL
serum using supportive liquid-liquid extraction and Diels-Alder
derivatization prior to LC-MS/MS analysis. Chromatographic
separations were achieved using Hypersil Gold C18 column
(150 ×2.1 mm; 1.9 µ) at flow rate 0.2 ml/min, operated in
Electrospray Ionization (ESI) positive mode and analyzed by
multiple reaction monitoring (MRM) method. The limit of
quantification (LOQ) for 1,25(OH)2D were 10 pg/mL, 3-epi-
25(OH)D, and 24,25(OH)2D were 50 pg/mL while 25(OH)D3
and 25(OH)D2 were 0.5 and 0.25 ng/mL, respectively. All
methods employed were performed in accordance with the
relevant guidelines and regulations.
Frontiers in Endocrinology | www.frontiersin.org 2January 2019 | Volume 10 | Article 13
8
Cunningham et al. Vitamin D and Embryo Development in PCOS
FIGURE 1 | 7-dehydrocholesterol in the skin is converted to previtamin D3 and then is thermally isomerize to vitamin D3. Transport of vitamin D3from the skin to the
liver is via the Vitamin D binding protein (DBP) transports 25 hydroxy vitamin D (25(OH)D3) to the kidney. 25(OH)D3/DBP is filtered by the glomeruli and 25(OH)D3is
taken up into the tubular cells, following DBP binding to megalin, a transmembrane protein. 25(OH)D3undergoes a second hydroxylation step by the
1-alpha-hydroxylase Cyp27B1, converting to the active 1α,25 (OH)2D3, whilst 24 hydroxylase Cyp27A1 converts to 24,25(OH)2D3. Keratinocytes contribute to the
3-epimerase activity converting 25(OH)D3to 3-epi-25(OH)D3, and 1α,25 (OH)2D3to 3-epi-1α,25 (OH)2D3, but the exact sites of activity remain unknown.
3epi25(OH)D3is equally converted by Cyp27A1 and Cyp27B1 as 25(OH)D3. *Vitamin D2derived from yeasts and fungi (mushrooms) is converted to 25(OH)D2in the
liver and to 1,25(OH)2D2and 24,25(OH)2D2in the kidney (adapted from 14, 16, and 21). It is unclear if the 3-epimers may be back converted.
All patients underwent a standard IVF antagonist protocol.
The patients commenced their rFSH stimulation on day 2 of their
menstrual cycle using either Merional (Pharmasure) or Gonal-F
(Merck Serono). A GnRH antagonist (Cetrotide: Merck Serono)
was used to prevent a premature LH surge.
The patients underwent ultrasound scans from day 7 to
observe the ovarian response to stimulation and were repeated
every 48 h. The scans were used to measure the diameters of
the follicles thus observing response and follicle numbers. Final
maturation was triggered when two or more leading follicles were
18 mm using human chorionic gonadotrophin [hCG, Pregnyl
(Merck Sharp and Dohme)].
Transcervical embryo transfer was performed and embryos
were classified using standard criteria (27) at the cleavage stage
(day 2–3 after egg collection) and for blastocyst stage (day 5–6
after egg collection). Top Quality embryos on Day 3 as per Alpha
Consensus (28). Embryo transfers were performed on either day
3 or ideally at day 5 (blastocyst) to give the best chance for
implantation as this timing is similar compared to natural cycle
embryos moving into the uterus.
Data Analysis and Statistics
Statistical analysis was performed using SPSS (v22, Chicago,
Illinios). Descriptive data is presented as mean ±SD for
continuous data and n (%) for categorical data. t-tests or
Mann Whitney tests were used to compare means/medians
where appropriate, and associations used Pearson’s correlation or
Spearman’s correlation as appropriate. A p<0.05 was considered
to indicate statistical significance. There was no comparative
study on which to base a formal power calculation; therefore,
power and sample size for a pilot study was performed (29);
therefore, to account for a minimum of 20 degrees-of-freedom
to estimate effect size and variability a minimum of 25 patients
per group were required to allow covariate adjustment.
RESULTS
Baseline characteristics of the 59 patients are shown in Table 1
where is can be seen that patients were non-obese, age, and
weight matched. There were significant differences in ovarian
reserve parameters antral follicle count (AFC) and anti-Mullerian
Hormone, (AMH), and androgen status between the groups,
however there was no significant difference in fasting insulin,
HOMA-IR or the vitamin D metabolites (Table 1).
There was a correlation between the levels of 25(OH)D3,
and embryo fertilization rates in PCOS patients (r=0.44; p
=0.03) that were not seen in the control group. However,
between the PCOS and control groups there were no differences
for any of the metabolic or embryo parameters for 25(OH)D2,
24R,25(OH)2D3, 1,25(OH)2D3, or 3epi25(OH)D3. When all of
Frontiers in Endocrinology | www.frontiersin.org 3January 2019 | Volume 10 | Article 13
9
Cunningham et al. Vitamin D and Embryo Development in PCOS
TABLE 1 | Mean demographics and biochemical data.
Control (n=30) PCOS (n=29) p-value
Mean data (±S.D.) Mean data (±S.D.)
Age 32.6 ±4.7 30.9 ±4.8 0.14
BMI 25.5 ±3.6 26.0 ±3.8 0.56
Menarche 13.0 ±2.0 13.0 ±1.1 0.99
Anovulatory 5 25 0.0001***
Duration of subfertility 3.9 ±1.8 3.4 ±1.6 0.84
Total antral follicle count 17.2 ±6.8 38.4 ±17.8 0.0001***
Fasting insulin (mIU/ml) 7.68 ±4.0 8.13 ±4.7 0.69
Fasting glucose (mmol/L) 4.81 ±0.4 4.62 ±0.4 0.06
HOMA-IR 1.71 ±1.0 1.72 ±1.0 0.97
SHBG 110.9 ±82.4 63.9 ±49.8 0.01*
Testosterone (mmol/L) 0.8 ±0.4 1.4 ±0.8 0.0004***
Free androgen index 1.35 ±0.6 4.21 ±2.9 0.0001***
25-hydroxyvitamin D3
(ng/mL)
46.2 ±23.5 54.0 ±27.4 0.24
25-hydroxyvitamin D2
(ng/ml)
0.5 ±0.3 0.6 ±0.5 0.73
1,25-dihydroxyvitamin D3 0.03 ±0.02 0.04 ±0.2 0.63
24R,25-dihydroxyvitamin
D3
0.8 ±0.5 1.3 ±0.6 0.003**
3-epi-25-hydroxyvitamin
D3
0.4 ±0.4 0.7 ±1.2 0.89
*p<0.01, **p<0.001, ***p<0.0001.
the subjects were combined there was a correlation between the
levels of 25(OH)D2and cleavage rate (r=0.31; p=0.05), G3D3
(r=0.40; p=0.009) and blastocyst (r=0.40; p=0.022);
there was a correlation between 3epi25(OH)D3and fertility rate
(r=0.33; p<0.04) and a negative correlation with HOMA-IR
(r= −0.33; p<0.02); 24R,25(0H)2D3correlated with AMH
(r=0.1; p=0.03) antral follicle count (r=0.2; p=0.003),
eggs retrieved (r=0.14; p=0.009) and G3D3 (r=0.22; p=
0.002), and negatively with HOMA-IR (r= −0.07; p<0.01).
There was no correlation of the active 1,25(0H)2D3with any of
the metabolic or embryo parameters.
There was a correlation between the levels of 25(OH)D3with
both 24R,25(OH)2D3and 3epi25(OH)D3(r=0.91, p<0.001; r
=0.35, p<0.015, respectively.
As a cohort, 25-hydroxyvitamin D levels were low did not
differ between the controls and the PCOS group. The Endocrine
Society defines vitamin D deficiency, insufficiency and replete
as (20 ng/mL, 20–30 ng/mL and 30 ng/mL, respectively (30)
that was reflected in controls and PCOS as, deficient, 51 vs. 41%;
insufficient, 33 vs. 35%; deficient, 10 vs. 24%.
IVF cycle characteristics are represented in Table 2 showing
that the PCOS group had significantly greater numbers of follicles
aspirated and eggs retrieved compared to the controls, and the
mean fertilization and cleavage rates were significantly higher for
the PCOS group, though embryos quality did not differ.
There was a significantly negative correlation between SHBG
and 25-hydroxyvitamin D3 in the PCOS subjects, however after
adjusting for BMI, SHBG was not significantly associated with
25-hydroxyvitamin D3.
TABLE 2 | Mean outcome data for stimulated ovarian cycle for Control and PCOS
groups. G3D3: Top Quality embryos on Day 3 as per Alpha Consensus (28).
Control (N=30) PCOS (N=28) p-value
Mean (±S.D.) Mean (±S.D.)
Endometrium at
oocyte retrieval
10.31 ±1.78 10.72 ±2.06 0.42
Follicles aspirated 11.47 ±5.11 15.96 ±5.30 0.002**
Eggs retrieved 8.47 ±5.08 11.29 ±5.02 0.04*
Fertilization 4.82 ±2.65 8.43 ±3.87 0.0003***
Cleavage 4.68 ±2.72 7.26 ±4.40 0.01*
G3D3 3.00 ±2.29 4.17 ±3.47 0.16
Blastocyst 1.46 ±1.77 2.91 ±3.01 0.05
PDT 11 10 0.86
Clinical pregnancy 10 7 0.24
*p<0.01, **p<0.001, ***p<0.0001.
DISCUSSION
This study has shown that 25(OH)D3was associated with
higher fertility rates in PCOS compared to non-obese, age, and
weight matched control subjects, but that this was not seen
for the other vitamin D metabolites. This was surprising given
that there was no difference in the 25(OH)D3levels between
the PCOS and control group; however, it is recognized that
ova in PCOS may be at a less mature stage compared to
normal and therefore there is a possibility that they may be
more 25-hydroxyvitamin D responsive to allow those ovum
within the stimulated follicles to reach a more mature stage
prior to ovum retrieval, resulting in a greater capability to
achieve fertilization. PCOS women typically produced more
poor quality oocytes, with lower fertilization, cleavage and
implantation rates (31,32). The impaired oocyte maturation
and resultant embryonic developmental competence in PCOS
women is possibly due to the abnormal endocrine/paracrine
functions and the environment within the follicle at the time of
folliculogenesis (33,34). No differences in the embryological data
for 25(OH)D3were found, in accord with others (25). It may
have been speculated that the active 1,25(OH)2D3may have a
greater influence on fertility at higher levels, but this was not seen
in this study, with no correlation with fertilization or embryo
data. Vitamin D is involved in the regulation of AMH and FSH
gene expression (31,35), and high dose 25(OH)D3has been
shown to increase serum AMH levels in vitamin D insufficiency
(36). In this study only the metabolite 24R,25(OH)2D3correlated
with AMH and antral follicle count; 24R,25(OH)2D3is an active
metabolite [It can be converted to 1,24,25-trihydroxyvitamin D3
through the C24 oxidation pathway (37)] as it has been shown
to induce non-genomic signaling pathways and suppresses Apo
A-1 in hep G cells (38), may have a physiological role in the
growth plate formation (14), therefore a direct effect on the
ovary cannot be excluded. However, 24,25 dihydroxyvitamin D
is associated with blood levels of 25-hydroxyvitamin D and given
that both were very significantly associated it is unlikely that
24R,25(OH)2D3was having a unique biological effect and indeed
was dependent on serum 25(OH)D3levels.
Frontiers in Endocrinology | www.frontiersin.org 4January 2019 | Volume 10 | Article 13
10
Cunningham et al. Vitamin D and Embryo Development in PCOS
There was a positive association for an increase in overall
fertility rate with the 3epi25(OH)D3that was not seen for the
other vitamin D metabolites. Little is known about the epimers
of vitamin D, with the assumption that they are biologically less
potent (21,23), but whilst that is likely for bone metabolism it
may not be the case for ovarian effects. Epimers are compounds
that have identical structure (and therefore identical molecular
weight) with the exception of a stereochemical difference at one
site. Other than the measurement of 3epi25(OH)D3to enable
the more accurate determination of 25(OH)D3, in children where
they have been shown to be higher, there has been little research
done on the C3 epimers (39) in order to know the significance
of this observation on fertility, but it is of interest to note
that serum lipids were discrepant for 25(OH)D epimeric forms
suggesting a differential effect (40). However, 3epi25(OH)D3is
associated with blood levels of 25-hydroxyvitamin D and given
that both were very significantly associated it is unlikely that
3epi25(OH)D3was having a unique biological effect and indeed
was dependent on serum 25(OH)D3levels.
Whole group analysis for 25(OH)D2, but not 25(OH)D3was
positively associated with increased cleavage rate, G3D3 and
blastocyst, though overall embryo quality did not differ. Whilst
vitamin D3 supplements are better than vitamin D2 to raise
vitamin D levels (41,42), their biological effects may not be
the same in different systems (43), and their differential effect
in the ovary needs to be clarified. The role of vitamin D in
fertilization remains controversial and the specific roles of 25-
hydroxyvitamin D levels largely unknown. Observational studies
have reported vitamin D levels within serum and follicular fluid
to be highly correlated and that those with higher serum and
follicular fluid levels of vitamin D had significantly higher clinical
pregnancy rates (12). Other studies have found no correlation
between serum and follicular fluid levels of vitamin D and
IVF outcomes (10,35,44). Conversely, two cohort studies
comparing serum vitamin D levels and pregnancy rates in women
undergoing fresh IVF showed that clinical pregnancy rates were
significantly lower in women who were vitamin D deficient
(24,25). Given the controversy, the implication of this data is
that in slim PCOS women undergoing IVF that their vitamin D
status should be determined, vitamin D replacement undertaken
for those deficient prior to IVF may be of benefit, or at least do
not harm, until future clarification becomes available.
These data suggest that vitamin D deficiency may not be a
homogeneous entity but rather may depend on the different
vitamin D metabolites present giving resultants effects, and
therefore may account for the heterogeneity and controversy
surrounding vitamin D deficiency effects and the response to
replacement (42,45). Whilst the effects of vitamin D and its
metabolites on bone and calcium metabolism are well-known,
vitamin D metabolite effects on other systems may not be
directly equipotent for each metabolite or comparable. For
example 1,25(OH)2-3-epi-D3may be biologically less active
than 1,25(OH)2D3, for increasing calcium, but may have
greater PTH suppression (39). However, the kidneys produce
1,25-dihydroxyvitamin D for regulating calcium and bone
metabolism (46). Therefore, measuring a blood level of 1,25-
dihydroxyvitamin D may not reflect its other biologic functions,
but rather local production of 1,25-dihydroxyvitamin D may
have its major benefit (47). Our observation that higher blood
levels of 25-hydroxyvitamin D are related to the outcome
measures could also be due to the higher substrate availability
for the local production of 1,25-dihydroxyvitamin D rather than
25-hydroxyvitamin D having a direct effect.
When weight and aged matched then there was no difference
in vitamin D metabolite levels between PCOS and normal
women. These results differ from previous studies that found
Vitamin D deficiency to be more common in PCOS subjects
(48); however, in this study the PCOS patients were specifically
non-obese and it is well-recognized that vitamin D levels fall
in obesity that may account for this observation (6). Negative
correlations with HOMA-IR were seen for both 24R,25(OH)2D3,
and 3epi25(OH)D3, but no association was seen for either
25(OH)D2or 25(OH)D3, suggesting that an association with
insulin resistance may depend on the metabolites present. There
were no correlations between testosterone or oocyte quality in the
PCOS patients in this study These observations are in accord with
some studies, though not with others (4,49); however, in those
reported studies patients were not intentionally weight matched.
High dose 25(OH)D3replacement was not associated with an
improvement in insulin sensitivity in PCOS subjects (50).
This study was specifically designed to look at the relationship
of vitamin D levels with fertilization rates and therefore was not
powered to look at pregnancy rates that would require a much
larger sample population. Furthermore, the overall sample size
is small and further work is needed, particularly to determine if
these findings are true for the differing PCOS phenotypes within
the Rotterdam criteria. However, the strengths of this study were
that the patients were age and weight matched from the same
ethnic background and that all of the vitamin D metabolites were
measured by state of the art methods.
In conclusion, non-obese age and weight matched PCOS
women showed that 25(OH)D3was associated with the
fertilization rate, compared to controls; however, vitamin D
metabolites were associated with embryology parameters and
HOMA-IR, suggesting a possible relationship between differing
vitamin D metabolites, oocyte maturation and insulin sensitivity
in non-obese PCOS patients.
AUTHOR CONTRIBUTIONS
TC was involved in the study design, acquisition of data, analysis
and interpretation of data, and paper drafting. HM, NA, and AL
were involved in vitamin D analysis and drafting the manuscript.
VA and SD were involved in analysis and interpretation of data,
and paper drafting. SA, EK, SM, and TS were involved in the
study design, supervision, paper drafting, and contributed to the
interpretation of the data. All authors read and approved the final
manuscript.
FUNDING
The publication of this article was funded by the Qatar National
Library.
ACKNOWLEDGMENTS
This work was part of an MD thesis and its publishing is in line
with the University of Hull policy, and can be accessed online
https://hydra.hull.ac.uk/resources/hull:14051 online (51).
Frontiers in Endocrinology | www.frontiersin.org 5January 2019 | Volume 10 | Article 13
11
Cunningham et al. Vitamin D and Embryo Development in PCOS
REFERENCES
1. Azziz R, Woods KS, Reyna R, Key TJ, Knochenhauer ES, Yildiz BO.
The prevalence and features of the polycystic ovary syndrome in an
unselected population. J Clin Endocrinol Metabol. (2004) 89:2745–9.
doi: 10.1210/jc.2003-032046
2. Franks S. Polycystic ovary syndrome. N Engl J Med. (1995) 333:853–61.
doi: 10.1056/NEJM199509283331307
3. Thomson RL, Spedding S, Buckley JD. Vitamin D in the aetiology and
management of polycystic ovary syndrome. Clin Endocrinol. (2012) 77:343–
50. doi: 10.1111/j.1365-2265.2012.04434.x
4. Ngo DT, Chan WP, Rajendran S, Heresztyn T, Amarasekera A, Sverdlov AL,
et al. Determinants of insulin responsiveness in young women: impact of
polycystic ovarian syndrome, nitric oxide, and vitamin D. Nitric Oxide (2011)
25:326–30. doi: 10.1016/j.niox.2011.06.005
5. Wehr E, Pilz S, Schweighofer N, Giuliani A, Kopera D, Pieber TR,
et al. Association of hypovitaminosis D with metabolic disturbances
in polycystic ovary syndrome. Eur J Endocrinol. (2009) 161:575–82.
doi: 10.1530/EJE-09-0432
6. Diamanti-Kandarakis E, Dunaif A. Insulin resistance and the polycystic ovary
syndrome revisited: an update on mechanisms and implications. Endocr Rev.
(2012) 33:981–1030. doi: 10.1210/er.2011-1034
7. Asemi Z, Foroozanfard F, Hashemi T, Bahmani F, Jamilian M, Esmaillzadeh
A. Calcium plus vitamin D supplementation affects glucose metabolism
and lipid concentrations in overweight and obese vitamin D deficient
women with polycystic ovary syndrome. Clin Nutr. (2015) 34:586–92.
doi: 10.1016/j.clnu.2014.09.015
8. Bacopoulou F, Kolias E, Efthymiou V, Antonopoulos CN, Charmandari E.
Vitamin D predictors in polycystic ovary syndrome: a meta-analysis. Eur J
Clin Invest. (2017) 47:746–55. doi: 10.1111/eci.12800
9. Rudick B, Ingles S, Chung K, Stanczyk F, Paulson R, Bendikson K.
Characterizing the influence of vitamin D levels on IVF outcomes. Hum
Reprod. (2012) 27:3321–7. doi: 10.1093/humrep/des280
10. Anifandis GM, Dafopoulos K, Messini CI, Chalvatzas N, Liakos N, Pournaras
S, et al. Prognostic value of follicular fluid 25-OH vitamin D and
glucose levels in the IVF outcome. Reprod Biol Endocrinol. (2010) 8:91.
doi: 10.1186/1477-7827-8-91
11. Rudick BJ, Ingles SA, Chung K, Stanczyk FZ, Paulson RJ,
Bendikson KA. Influence of vitamin D levels on in vitro fertilization
outcomes in donor-recipient cycles. Fertil Steril. (2014) 101:447–52.
doi: 10.1016/j.fertnstert.2013.10.008
12. Ozkan S, Jindal S, Greenseid K, Shu J, Zeitlian G, Hickmon C, et al. Replete
vitamin D stores predict reproductive success following in vitro fertilization.
Fertil. Steril. (2010) 94:1314–9. doi: 10.1016/j.fertnstert.2009.05.019
13. Wortsman J, Matsuoka LY, Chen TC, Lu Z, Holick MF. Decreased
bioavailability of vitamin D in obesity. Am J Clin Nutr. (2000) 72:690–3.
doi: 10.1093/ajcn/72.3.690
14. Bikle DD. Vitamin D metabolism, mechanism of action,
and clinical applications. Chem Biol. (2014) 21:319–29.
doi: 10.1016/j.chembiol.2013.12.016
15. Sakaki T, Kagawa N, Yamamoto K, Inouye K. Metabolism of vitamin D3 by
cytochromes P450. Front Biosci. (2005) 10:119–34. doi: 10.2741/1514
16. Christakos S, Dhawan P, Verstuyf A, Verlinden L, Carmeliet G. Vitamin D:
metabolism, molecular mechanism of action, and pleiotropic effects. Physiol
Rev. (2016) 96:365–408. doi: 10.1152/physrev.00014.2015
17. Adams JS, Rafison B, Witzel S, Reyes RE, Shieh A, Chun R, et al. Regulation
of the extrarenal CYP27B1-hydroxylase. J Steroid Biochem Mol Biol. (2014)
144 Pt A:22–7. doi: 10.1016/j.jsbmb.2013.12.009
18. Lösel R, Wehling M. Nongenomic actions of steroid hormones.
Nat Rev Mol Cell Biol. (2003) 4:46–55. doi: 10.1038/
nrm1009
19. Agic A, Xu H, Altgassen C, Noack F, Wolfler MM, Diedrich K, et al.
Relative expression of 1,25-dihydroxyvitamin D3 receptor, vitamin D 1
alpha-hydroxylase, vitamin D 24-hydroxylase, and vitamin D 25-hydroxylase
in endometriosis and gynecologic cancers. Reprod Sci. (2007) 14:486–97.
doi: 10.1177/1933719107304565
20. Viganò P, Lattuada D, Mangioni S, Ermellino L, Vignali M, Caporizzo
E, et al. Cycling and early pregnant endometrium as a site of regulated
expression of the vitamin D system. J Mol Endocrinol. (2006) 36:415–24.
doi: 10.1677/jme.1.01946
21. Kamao M, Tatematsu S, Hatakeyama S, Sakaki T, Sawada N, Inouye K, et al.
C-3 epimerization of vitamin D3 metabolites and further metabolism of C-
3 epimers: 25-hydroxyvitamin D3 is metabolized to 3-epi-25-hydroxyvitamin
D3 and subsequently metabolized through C-1alpha or C-24 hydroxylation. J
Biol Chem. (2004) 279:15897–907. doi: 10.1074/jbc.M311473200
22. Schleicher RL, Encisco SE, Chaudhary-Webb M, Paliakov E, McCoy LF,
Pfeiffer CM. Isotope dilution ultra performance liquid chromatography-
tandem mass spectrometry method for simultaneous measurement
of 25-hydroxyvitamin D2, 25-hydroxyvitamin D3 and 3-epi-25-
hydroxyvitamin D3 in human serum. Clin Chim Acta (2011) 412:1594–9.
doi: 10.1016/j.cca.2011.05.010
23. Molnár F, Sigüeiro R, Sato Y, Araujo C, Schuster I, Antony P, et al.
1alpha,25(OH)2-3-epi-vitamin D3, a natural physiological metabolite of
vitamin D3: its synthesis, biological activity and crystal structure with
its receptor. PLoS ONE (2011) 6:e18124. doi: 10.1371/journal.pone.00
18124
24. Garbedian K, Boggild M, Moody J, Liu KE. Effect of vitamin D status on
clinical pregnancy rates following in vitro fertilization. CMAJ Open (2013)
1:E77–82. doi: 10.9778/cmajo.20120032
25. Polyzos NP, Anckaert E, Guzman L, Schiettecatte J, Van Landuyt L, Camus M,
et al. Vitamin D deficiency and pregnancy rates in women undergoing single
embryo, blastocyst stage, transfer (SET) for IVF/ICSI. Hum Reprod. (2014)
29:2032–40. doi: 10.1093/humrep/deu156
26. Rotterdam ESHRE/ASRM-Sponsored PCOS Consensus Workshop Group.
Revised 2003 consensus on diagnostic criteria and long-term health risks
related to polycystic ovary syndrome (PCOS). Hum Reprod. (2004) 19:41–7.
doi: 10.1093/humrep/deh098
27. Cutting R, Morroll D, Roberts SA, Pickering S, Rutherford A. Elective
single embryo transfer: guidelines for practice British Fertility Society
and Association of Clinical Embryologists. Hum Fertil. (2008) 11:131–46.
doi: 10.1080/14647270802302629
28. ALPHA Scientists in Reproductive Medicine, ESHRE Special Interest
Group Embryology. Istanbul consensus workshop on embryo assessment:
proceedings of an expert meeting. Reprod Biomed Online (2011) 22:632–46.
doi: 10.1016/j.rbmo.2011.02.001
29. Birkett MA, Day SJ. Internal pilot studies for estimating sample size. Stat Med.
(1994) 13:2455–63. doi: 10.1002/sim.4780132309
30. Holick MF, Binkley NC, Bischoff-Ferrari HA, Gordon CM, Hanley DA,
Heaney RP, et al. Evaluation, treatment, and prevention of vitamin D
deficiency: an Endocrine Society clinical practice guideline. J Clin Endocrinol
Metab. (2011) 96:1911–30. doi: 10.1210/jc.2011-0385
31. Irani M, Merhi Z. Role of vitamin D in ovarian physiology and its implication
in reproduction: a systematic review. Fertil Steril. (2014) 102:460–8.e463.
doi: 10.1016/j.fertnstert.2014.04.046
32. Sahu B, Ozturk O, Ranierri M, Serhal P. Comparison of oocyte quality and
intracytoplasmic sperm injection outcome in women with isolated polycystic
ovaries or polycystic ovarian syndrome. Arch Gynecol Obstet. (2008) 277:239–
44. doi: 10.1007/s00404-007-0462-x
33. Barber TM, Golding SJ, Alvey C, Wass JA, Karpe F, Franks S, et al. Global
adiposity rather than abnormal regional fat distribution characterizes women
with polycystic ovary syndrome. J Clin Endocrinol Metab. (2008) 93:999–1004.
doi: 10.1210/jc.2007-2117
34. Wood JR, Dumesic DA, Abbott DH, Strauss JF. Molecular abnormalities in
oocytes from women with polycystic ovary syndrome revealed by microarray
analysis. J Clin Endocrinol Metab. (2007) 92:705–13. doi: 10.1210/jc.
2006-2123
35. Firouzabadi RD, Rahmani E, Rahsepar M, Firouzabadi MM. Value of
follicular fluid vitamin D in predicting the pregnancy rate in an IVF
program. Arch Gynecol Obstet. (2014) 289:201–6. doi: 10.1007/s00404-013-
2959-9
36. Dennis NA, Houghton LA, Pankhurst MW, Harper MJ, McLennan IS. Acute
supplementation with high dose vitamin D3 increases serum anti-mullerian
hormone in young women. Nutrients (2017) 9:E719. doi: 10.3390/nu9070719
37. Tieu EW, Tang EK, Tuckey RC. Kinetic analysis of human CYP24A1
metabolism of vitamin D via the C24-oxidation pathway. FEBS J. (2014)
281:3280–96. doi: 10.1111/febs.12862
Frontiers in Endocrinology | www.frontiersin.org 6January 2019 | Volume 10 | Article 13
12
Cunningham et al. Vitamin D and Embryo Development in PCOS
38. Wehmeier K, Onstead-Haas LM, Wong NC, Mooradian AD, Haas
MJ. Pro-inflammatory signaling by 24,25-dihydroxyvitamin D3 in
HepG2 cells. J Mol Endocrinol. (2016) 57:87–96. doi: 10.1530/JME-
16-0009
39. Lensmeyer G, Poquette M, Wiebe D, Binkley N. The C-3 epimer of 25-
hydroxyvitamin D is present in adult serum. J Clin Endocrinol Metab. (2012)
97:163–8. doi: 10.1210/jc.2011-0584
40. Chailurkit LO, Aekplakorn W, Srijaruskul K, Ongphiphadhanakul B.
Discrepant association of serum C-3 epimer of 25-hydroxyvitamin
D versus non-epimeric 25-hydroxyvitamin D with serum lipid
levels. Lipids Health Dis. (2016) 15:157. doi: 10.1186/s12944-016-
0333-1
41. Tripkovic L, Lambert H, Hart K, Smith CP, Bucca G, Penson S,
et al. Comparison of vitamin D2 and vitamin D3 supplementation
in raising serum 25-hydroxyvitamin D status: a systematic review and
meta-analysis. Am J Clin Nutr. (2012) 95:1357–64. doi: 10.3945/ajcn.111.
031070
42. Hammami MM, Yusuf A. Differential effects of vitamin D2 and D3
supplements on 25-hydroxyvitamin D level are dose, sex, and time dependent:
a randomized controlled trial. BMC Endocrine Disorders (2017) 17:12.
doi: 10.1186/s12902-017-0163-9
43. Chabas JF, Stephan D, Marqueste T, Garcia S, Lavaut MN, Nguyen C,
et al. Cholecalciferol (vitamin D) improves myelination and recovery
after nerve injury. PLoS ONE (2013) 8:e65034. doi: 10.1371/journal.pone.0
065034
44. Aleyasin A, Hosseini MA, Mahdavi A, Safdarian L, Fallahi P, Mohajeri MR,
et al. Predictive value of the level of vitamin D in follicular fluid on the
outcome of assisted reproductive technology. Eur J Obstet Gynecol Reprod
Biol. (2011) 159:132–7. doi: 10.1016/j.ejogrb.2011.07.006
45. Al Mheid I, Quyyumi AA. Vitamin D and cardiovascular disease:
controversy unresolved. J Am Coll Cardiol. (2017) 70:89–100.
doi: 10.1016/j.jacc.2017.05.031
46. Adams JS, Ramin J, Rafison B, Windon C, Windon A, Liu PT. Redefining
human vitamin D sufficiency: back to the basics. Bone Res. (2013) 1:2–10.
doi: 10.4248/BR201301002
47. Adams IP, Dack S, Dickinson FM, Ratledge C. The distinctiveness of
ATP:citrate lyase from Aspergillus nidulans.Biochim Biophys Acta (2002)
1597:36–41. doi: 10.1016/S0167-4838(02)00276-5
48. Mahmoudi T, Gourabi H, Ashrafi M, Yazdi RS, Ezabadi Z. Calciotropic
hormones, insulin resistance, and the polycystic ovary syndrome. Fertil Steril.
(2010) 93:1208–14. doi: 10.1016/j.fertnstert.2008.11.031
49. Hahn S, Haselhorst U, Tan S, Quadbeck B, Schmidt M, Roesler S, et al.
Low serum 25-hydroxyvitamin D concentrations are associated with insulin
resistance and obesity in women with polycystic ovary syndrome. Exp Clin
Endocrinol Diabetes (2006) 114:577–83. doi: 10.1055/s-2006-948308
50. Raja-Khan N, Shah J, Stetter CM, Lott ME, Kunselman AR, Dodson WC, et al.
High-dose vitamin D supplementation and measures of insulin sensitivity in
polycystic ovary syndrome: a randomized, controlled pilot trial. Fertil Steril.
(2014) 101:1740–6. doi: 10.1016/j.fertnstert.2014.02.021
51. Cunningham TC. The Effect of Environmental, Physical, and Nutritional
Factors on in vitro Fertilisation. University of Hull (2016).
Conflict of Interest Statement: The authors declare that the research was
conducted in the absence of any commercial or financial relationships that could
be construed as a potential conflict of interest.
Copyright © 2019 Cunningham, Allgar, Dargham, Kilpatrick, Sathyapalan,
Maguiness, Mokhtar Rudin, Abdul Ghani, Latiff and Atkin. This is an open-access
article distributed under the terms of the Creative Commons Attribution License (CC
BY). The use, distribution or reproduction in other forums is permitted, provided
the original author(s) and the copyright owner(s) are credited and that the original
publication in this journal is cited, in accordance with accepted academic practice.
No use, distribution or reproduction is permitted which does not comply with these
terms.
Frontiers in Endocrinology | www.frontiersin.org 7January 2019 | Volume 10 | Article 13
13
REVIEW
published: 19 February 2019
doi: 10.3389/fendo.2019.00079
Frontiers in Endocrinology | www.frontiersin.org 1February 2019 | Volume 10 | Article 79
Edited by:
Daniel David Bikle,
University of California, San Francisco,
United States
Reviewed by:
Roger Bouillon,
Katholieke Universiteit Leuven,
Belgium
Jan Josef Stepan,
Charles University, Czechia
*Correspondence:
Rolf Jorde
rolf.jorde@unn.no
Specialty section:
This article was submitted to
Bone Research,
a section of the journal
Frontiers in Endocrinology
Received: 21 December 2018
Accepted: 30 January 2019
Published: 19 February 2019
Citation:
Jorde R (2019) The Role of Vitamin D
Binding Protein, Total and Free
25-Hydroxyvitamin D in Diabetes.
Front. Endocrinol. 10:79.
doi: 10.3389/fendo.2019.00079
The Role of Vitamin D Binding
Protein, Total and Free
25-Hydroxyvitamin D in Diabetes
Rolf Jorde 1,2
*
1Tromsø Endocrine Research Group, Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway,
2Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
Vitamin D is important for bone health, but may also have extra-skeletal effects. Vitamin D
and its binding protein DBP have immunological effects and may therefore be important
in the development of type 1 diabetes (T1DM), and low serum levels of 25-hydroxyvitamin
D (25(OH)D) are associated with later development of type 2 diabetes (T2DM). However,
it has so far been difficult to convincingly show an effect of vitamin D supplementation
on prevention or treatment of diabetes. The serum level of 25(OH)D has traditionally
been used as a marker of a subject’s vitamin D status. This measurement includes both
25(OH)D bound to DBP and albumin as well as the free from of 25(OH)D. However,
according to the free hormone hypothesis, the free form is the biologically active.
Previously the free form of 25(OH)D had to be calculated based on measurements
of 25(OH)D, DBP, and albumin, but recently a method for direct measurement of free
25(OH)D has become commercially available. This is important in clinical conditions
where the amount of DBP is affected, and has caused a renewed interest in which vitamin
D metabolite to measure in clinical situations. In the present review the relations between
DBP, total and free 25(OH)D in T1DM and T2DM are described.
Keywords: diabetes, free vitamin D, vitamin D binding protein (DBP), single nucelotide polymorphisms, 25- hydroxy
vitamin D
INTRODUCTION
Vitamin D is produced in the skin upon UV-B exposure and is obtained through the diet where
fatty fish is the main source. Regardless of how it is obtained, vitamin D has to be hydroxylated
first in the liver to 25-hydroxyvitamin D (25(OH)D) and then in the kidneys to the active form
1,25-dihydroxyvitamin D (1,25(OH)2D) (1). These hydroxylations may also occur in peripheral
tissues (2).
In the circulation the major part of vitamin D, 25(OH)D and 1,25(OH)2D are bound to the
vitamin D binding protein (DBP), and to a lesser extent also to albumin. Only a small fraction
circulates in the free form (3). To exert their action, the vitamin D metabolites have to cross the cell
membrane into the cell [and for vitamin D and 25(OH)D also to be hydroxylated], where the active
form 1,25(OH)2D connects to the nuclear vitamin D receptor (VDR) (1).
14
Jorde DBP, Total and Free 25(OH)D and Diabetes
The endocytic receptors megalin and cubulin are present in
the renal tubuli and parathyroid cells (4), and at least in the
kidney enable transportation of the DBP-vitamin D complexes
into the cells (5). In other (and perhaps most) cell types, the
vitamin D metabolites have to pass the cell membranes in their
free un-bound form by passive diffusion (6).
The serum concentrations of vitamin D and 25(OH)D are
>100 times that of 1,25(OH)2D, and the DBP binding coefficients
as well as the potential for passive diffusion through cell
membranes differ between these vitamin D metabolites (6).
Accordingly, it is difficult to say which vitamin D metabolite,
or vitamin D metabolite-DBP complex is quantitatively the
most important for VDR activation and the one that should be
measured for evaluation of a subject’s vitamin D status (6,7).
For this purpose, one has traditionally measured the serum
25(OH)D level, since this metabolite is abundant, easy to
measure, and has a long half-life and therefore stable levels.
Furthermore, the hydroxylation from vitamin D to 25(OH)D is
substrate driven and the serum 25(OH)D level correlates strongly
with sun exposure and vitamin D intake and also correlates with
known vitamin D effects, like the suppression of the parathyroid
hormone (PTH) secretion (1).
The serum 25(OH)D that is measured is the total 25(OH)D,
which includes the DBP and albumin bound 25(OH)D as well
as the free form. Since the major part of 25(OH)D is bound
to DBP, the concentration of total 25(OH)D will depend on
the serum DBP concentration. The DBP concentration is fairly
stable throughout life, but increases with pregnancy and estrogen
supplementation. DBP is synthesized in the liver and accordingly
the serum DBP concentration is reduced in liver failure as well
as in malnutrition (8,9). Loss of proteins in the urine (like in
some subjects with diabetes) may also cause low serum DBP
levels (10,11). Thus, in situations with high serum DBP levels
like pregnancy, an even larger portion of the total 25(OH)D in
plasma is bound to DBP and accordingly the free form is reduced.
Conversely, in patients with liver cirrhosis where the serum level
of DBP is low, the free fraction is increased. Although there
is a strong correlation between total and free 25(OH)D (12),
measurement of total 25(OH)D may therefore not always reflect
the free form.
According to the free hormone hypothesis, it is the free form
of the hormone, which easily diffuses through cell membranes,
that is the biologically active, and the one to be measured (13).
This is exemplified for thyroid hormones, where the serum
concentration of tree thyroxine is regulated in a negative feedback
manner by the secretion of thyroid stimulating hormone (TSH).
In this system, changes in the concentration of thyroid hormone
binding globulin (TBG) will be compensated by increased or
decreased secretion of TSH keeping the free concentration of
thyroxine stable (14). This demonstrates the utility of the free
hormone concept for thyroid hormones.
This concept does not necessarily apply to the vitamin
D system where the active hormone 1,25(OH)2D can be
transported into (at least some) cells in a DBP-complex,
and also have its activating hydroxylations intracellularly.
Furthermore, 25(OH)D is in essence a pro-hormone not
regulated by negative feed-back control. Changes in DBP will
not induce changes in the hydroxylation of vitamin D to
25(OH)D since this is a substrate driven process. Increased
serum 25(OH)D concentrations may be accompanied by an
increased level of FGF-23, increased CYP24A1 expression and 24-
hydroxylase activity, and accelerated degradation of 25(OH)D to
24,25(OH)2D (15). However, this mechanism must for 25(OH)D
be of minor importance since the increase in free 25(OH)D
and total 25(OH)D after vitamin D supplementation is, at least
until serum 25(OH)D levels of approximately 150 nmol/L, quite
linear (12). Therefore, whether the total or the free form of
25(OH)D is the best vitamin D parameter cannot be decided on
theoretical grounds only, but has to be tested in clinical situations
as well (16,17).
There are many single nucleotide polymorphisms (SNPs) in
the DBP gene (GC gene, globulin–complex gene). Combinations
of two of these (rs7041 and rs4588) result in three polymorphic
alleles and six major phenotypes. These phenotypes may have
different binding affinities for the vitamin D metabolites (18)
and the serum 25(OH)D levels do differ between subjects with
different DBP phenotypes (12). The distribution of the six
variants also differs between races (19).
In addition to the skeleton vitamin D deficiency has
been associated with a number of diseases, like mortality,
cancer, immunological diseases, cardio-vascular diseases,
and diabetes (20). Most of these relations are based on
observational studies only, where 25(OH)D has been measured
in old serum samples and subsequent diseases recorded.
For these studies, measurement of total serum 25(OH)D
has been employed, whereas there has been little focus on
DBP [where the major part of the circulating 25(OH)D
is bound] or the free form which potentially may be the
most important.
The serum level of free 25(OH)D has traditionally been
calculated based on measurements of total 25(OH)D, DBP,
and albumin concentrations (2123). However, measurement
of DBP depends on type of antibody employed (monoclonal
or polyclonal) (19), and it has usually been assumed that the
vitamin D binding-coefficient for each of the six prevalent
DBP phenotypes are equal. The validity of the free 25(OH)D
calculations have therefore been questioned (24). Lately, kits for
direct measurement free 25(OH)D has become commercially
available which has caused a renewed interest in the relation
between free serum 25(OH)D, as well as DBP, and disease states
(25). However, further validation and standardization of this
assay is still needed in subjects with major illnesses or with
abnormal DBP or protein concentrations (16).
In the present review these relations will be summarized for
the metabolic disorders type 1 and type 2 diabetes (T1DM and
T2DM), presented separately.
T2DM
Serum 25(OH)D and T2DM
There are many reasons for why vitamin D could influence
the development of T2DM. Thus, the vitamin D activating
hydroxylases and the VDR are found in the pancreatic beta-
cells (26,27), 1,25(OH)2D may induce insulin secretion (28), and
Frontiers in Endocrinology | www.frontiersin.org 2February 2019 | Volume 10 | Article 79
15
Jorde DBP, Total and Free 25(OH)D and Diabetes
vitamin D may have an anti-inflammatory effect that may prevent
insulin resistance (29).
In line with this, there are a number of observational studies
on the relation between serum 25(OH)D concentration and
incident diabetes, and practically all confirm an association
(30). Thus, in a study by Afzal et al. on 31,040 subjects with
measurement of serum 25(OH)D followed for up to 34 years,
participants who had a 20 nmol/L reduction in 25(OH)D had a
16% increased risk of T2DM (31). Similarly, Ye et al. combined
22 studies in a meta-analysis that included 8,492 cases and 89,698
controls and found a 21% increased risk of T2DM per 25 nmol/L
lower 25(OH)D concentration (32).
However, for vitamin D there is a strong possibility of revers
causation and other methods than observational studies are
needed for confirmation, as recently reviewed by Angelotti and
Pittas (30).
There are a few RCTs with vitamin D specifically designed
for prevention of T2DM in subjects at risk. Thus, Davidson
et al. included 109 subjects with prediabetes and randomized
them to high dose vitamin D (mean weekly dose 88,865 IU)
vs. placebo. However, no significant effects on insulin secretion,
insulin sensitivity or development of diabetes were found after
1 year (33). Similarly, in a study from Tromsø, Norway,
Jorde et al. randomized 511 subjects with reduced glucose
tolerance to 20,000 IU vitamin D per week vs. placebo for
a maximum of 5 years, but found no difference between the
groups in development of T2DM (34). However, both studies
were underpowered for detection of minor effects. And finally,
the effect of giving vitamin D to subjects with established
T2DM do at best show a marginal effect on HbA1c with a
reduction of 0.32% in HbA1c as compared with placebo according
to a review by Lee et al. that included nine trial with 3,324
participants (35).
Another approach to the vitamin D—T2DM question is
the Mendelian randomization. Several SNPs are associated
with serum 25(OH)D level; SNPs in the DHCR7 gene
related to vitamin D synthesis, the CYP2R1 gene related
to 25-hydroxylation, and the CYP24A1 gene related to 24-
hydroxylation and degradation (36). When these SNPs are
combined to a genetic score, the highest vs. the lowest
scores result in 5–20% difference in serum 25(OH)D levels.
However, in the most recent and largest meta-analysis including
five studies with 28,144 cases and 76,344 non-cases, no
significant association with T2DM was found, neither for
the individual SNPs tested, nor when combined to a genetic
score (32).
There are, however, many shortcomings of the Mendelian
randomization approach. So far it only predicts differences in
serum 25(OH)D concentration and not the free fraction, and the
alleles tested only explain a small part of the variance in serum
25(OH)D level.
One may therefore conclude that although a low serum
25(OH)D level do predict development of T2DM, this is most
likely due to confounding or reverse causality, although minor
effects cannot be excluded. Hopefully the ongoing D2d study that
has included 2,423 participants with prediabets randomized to
4000 IU vitamin D daily vs. placebo may settle this question (37).
Free 25(OH)D and T2DM
There are several reports where the directly measured free
fraction of 25(OH)D has been compared with total 25(OH)D
regarding biological effects of vitamin D. Thus, Johnsen et al.
found a better correlation for free than for total 25(OH)D
regarding bone density (24), whereas that was not found in study
by Michaelsson et al. (38). For PTH similar relations have been
found for free and total 25(OH)D in most studies (24,3941),
whereas Lopez-Molina et al. in healthy children found better
correlation with markers of phosphocalcic metabolism for free
than for total 25(OH)D (42). Shieh et al. found in the early phase
(first 4 weeks) of vitamin D treatment the free 25(OH)D, but not
the total 25(OH)D, to be associated with a decrease in serum PTH
(43). In inflammatory diseases the results are also mixed with
free 25(OH)D correlating better to disease activity in ulcerative
colitis (44), whereas total 25(OH)D correlates best to activity in
systemic lupus erythematosus (45). For markers of inflammation
(IL-6) in older men free and total 25(OH)D appear to correlate
equally (46). And finally and most important, in a study by Yu
et al. the free but not total 25(OH)D was associated with risk
of mortality in patients with coronary artery disease (47). The
study included 1,387 patients followed for a median time of
6.7 years, during which period 205 patients died. The all-cause
mortality was 64% higher in the lowest free 25(OH)D quartile
vs. the highest free 25(OH)D quartile, whereas the corresponding
analysis using 25(OH)D did not show a significant difference or
trend across the quartiles.
So far, there are no studies where the free 25(OH)D has been
compared with total 25(OH)D as predictor for development of
T2DM. However, there is a publication by Lee et al. that included
1,189 non-diabetic subjects where the free and total form of
25(OH)D were measured and related to acute insulin response
and glucose disposition index based on intravenous glucose
tolerance tests (48). Both free and total 25(OH)D were positively
associated with these measures, but after adjustment for BMI,
only free 25(OH)D was significant related to insulin secretion.
Based on the above papers, one cannot conclude that
measurements of free vs. total serum 25(OH)D has any advantage
regarding vitamin D responses. This is also difficult to decide,
as comparisons of P-values and correlation coefficients give
indications only.
DBP and T2DM
In addition to being the carrier protein for vitamin D and its
metabolites, DBP has a number of other effects. It acts as a
carrier for free fatty acids (49), it binds actin and may prevent
actin polymerization during tissue damage (50,51), may act as a
macrophage activator and play a part in the inflammation process
by influencing the T-cell response (52). These immunological
effects may differ between the phenotypes (53), and the level of
DBP as well as the different DBP phenotypes might therefore at
least theoretically affect the development of not only T1DM (see
later) but also T2DM.
However, in a case-cohort study design with 958 cases and
3,489 controls Jorde et al. found no association between DBP
phenotypes (based on genotyping of rs4588 and rs7041) and
incident T2DM (54). Furthermore, there were no relations
Frontiers in Endocrinology | www.frontiersin.org 3February 2019 | Volume 10 | Article 79
16
Jorde DBP, Total and Free 25(OH)D and Diabetes
between the DBP phenotypes and lipids and blood pressure, but
a slight relation to hip circumference.
Prior to our study Wang et al. made a meta-analysis on
DBP SNPs and T2DM that included six studies (three Caucasian
and three Asian cohorts) with 1,191 cases and 882 controls. No
overall association between the DBP SNPs rs4588 and rs7041 and
T2DM was found. However, when analyzing the Asian cohorts
separately, there were significant associations with T2DM for
both rs7041 and rs4588 (55).
Also after the meta-analysis by Wang et al., Ye at al.
meta-analyzed the DBP SNP rs4588 regarding T2DM in
European cohorts including 28,144 cases and 76,344 controls.
A strong relation between rs4588 and serum 25(OH)D was
found, but not with T2DM (OR 1.00 (CI, 0.97 1.03)
(32). Accordingly, at least in Caucasians there appears to
be no relation between DBP phenotypes and development
of diabetes.
To the author’s knowledge, there are no longitudinal studies
regarding serum levels of DBP and T2DM. However, there is
one cross-sectional study by Leong et al. on 2,122 adult subjects
that included 201 with diabetes (56). The effect estimate per 50
mg/L DBP increase was 0.79 (95% CI 0.65–0.96) for diabetes,
and there was a marginal relation between higher DBP and lower
fasting blood glucose levels. However, as a cross-sectional study
it could not examine the impact of biological variability of DBP
over time.
T1DM
Serum 25(OH)D and T1DM
The 1α-hydroxylase, necessary for activation of vitamin D, is
expressed in immune cells like the B- and T-cells and the
antigen presenting cells (2). These cells may therefore synthesize
active vitamin D locally. Vitamin D has immune-modulatory
effects (57), and since T1DM is an autoimmune disorder, a
role for vitamin D in pathogenesis as well as treatment thus
possible (58).
However, in a study by Thorsen et al. using a case-cohort
design that included 459 children with T1DM and a control
group of 1,561, no association between maternal serum 25(OH)D
levels sampled repeatedly during pregnancy and subsequent
T1DM in the offsprings was found (59). Furthermore, in two
large Danish populations, one case-cohort study with 912 cases
and 2,866 controls followed for a maximum of 31 years and
a case-control study with 527 matched pairs followed for a
maximum of 23 years, Jacobsen et al. found no relation between
neonatal vitamin D status and later risk of T1DM (60). On the
other hand, there might be a link between intake of vitamin D in
childhood and development of T1DM as reported by Hyppönen
et al. in a birth-cohort study with 12,055 pregnant women in
northern Finland (61). This was also the conclusion in a meta-
analysis by Dong et al. from 2013 that included eight studies
(six case-control and two cohort studies) with vitamin D intake
during early life where the pooled OR for T1DM was 0.71 (95%
CI, 0.51–0.98) (62).
Furthermore, the serum levels of 25(OH)D are lower in
subjects with newly diagnosed T1DM (63) as well as later in the
course of disease compared to similarly aged subjects (64). There
may also be a beneficial effect by vitamin D supplementation
in newly diagnosed T1DM. This was reviewed by Gregoriou
et al. (65) who found a positive effects on the daily insulin dose,
fasting, and stimulated C-peptide response by vitamin D in two
studies. However, only 67 patients were randomized and the
effect was marginal.
To the author’s knowledge there are no studies reporting free
25(OH)D levels in T1DM.
DBP and T1DM
Since the immunological effects of DBP may differ between the
DBP phenotypes (53), relations between the DBP SNPs rs4588
and rs7041 and T1DM are of interest. This was reviewed by
Penna-Martinez and Badenhoop who found that in the majority
of the studies there was no relation between these SNPs and
T1DM (66). As an example, Cooper et al. who included 720
cases and 2,610 controls and used a Mendelian randomization
approach, found no relation between rs4588 and T1DM (67),
whereas in the two studies that did find an association with rs7041
the total number of cases was only 154 (68,69).
There are a few cross-sectional reports on serum DBP levels
in patients with T1DM. In a study by Blanton et al. that
included 203 subjects with T1DM and 153 controls, the serum
DBP levels were 10% lower in the T1DM patients (70). A
similar result was found by Thraikill et al. but they could for
a large part ascribe this to increased urinary loss of DBP in
the urine (11). Low serum DBP levels have also been described
in diabetic BB rats together with low serum 1,25(OH)2D
levels accompanied with reduced duodenal calcium absorption,
indicating the possible physiological importance of urinary DBP
loss (71).
CONCLUSIONS
For preventing or treating diabetes, the majority of clinical
studies do not indicate a major role for vitamin D
supplementation, with a possible exception for T1DM in
children. As for many other presumed extra-skeletal effects
of vitamin D, the effect on glucose metabolism must be small
(if present at all) and accordingly difficult to demonstrate. In
most of the vitamin D RCTs the results are also hampered by
the inclusion of subjects who are not truly vitamin D deficient
(72). However, since such subjects (and in particular young
children) need vitamin D for bone health, there are many ethical
problems in including vitamin D deficient subjects in long lasting
RCTs. The “perfect” vitamin D RCT will therefore probably not
be performed.
However, regarding vitamin D and health, the two crucial
questions are how much vitamin D we need for skeletal
health (which everyone agrees is vitamin D dependent),
and if supplementation above that will give any additional
health benefits.
So far, there are too few studies on the relative importance
of measuring total or free 25(OH)D in diabetes and glucose
Frontiers in Endocrinology | www.frontiersin.org 4February 2019 | Volume 10 | Article 79
17
Jorde DBP, Total and Free 25(OH)D and Diabetes
metabolism, and too few studies on the importance of DBP
concentration on development and progression of diabetes, to
draw firm conclusion. However, since it is difficult to show
an effect of vitamin D supplementation regarding diabetes, it
follows that finding the right form or metabolite of vitamin D
to measure (7), may for diabetes simply be a search for another
biomarker (73). In disease states with clearly altered DBP levels,
like pregnancy and liver cirrhosis, the situation obviously is
different (9).
AUTHOR CONTRIBUTIONS
The author confirms being the sole contributor of this work and
approved it for publication.
ACKNOWLEDGMENTS
UiT The Arctic University of Norway is gratefully acknowledged
for their support.
REFERENCES
1. DeLuca HF. Overview of general physiologic features and functions of vitamin
D. Am J Clin Nutr. (2004) 80:1689S96S. doi: 10.1093/ajcn/80.6.1689S
2. Bikle D. Nonclassic actions of vitamin D. J Clin Endocrinol Metab. (2009)
94:26–34. doi: 10.1210/jc.2008-1454
3. Christakos S, Dhawan P, Verstuyf A, Verlinden L, Carmeliet G. Vitamin D:
metabolism, molecular mechanism of action, and pleiotropic effects. Physiol
Rev. (2016) 96:365–408. doi: 10.1152/physrev.00014.2015
4. Lundgren S, Carling T, Hjälm G, Juhlin C, Rastad J, Pihlgren U, et al. Tissue
distribution of human gp330/megalin, a putative Ca2+-sensing protein. J
Histochem Cytochem. (1997) 45:383–92. doi: 10.1177/002215549704500306
5. Nykjaer A, Dragun D, Walther D, Vorum H, Jacobsen C, Herz
J, et al. An endocytic pathway essential for renal uptake and
activation of the steroid 25-(OH) vitamin D3. Cell (1999) 96:507–15.
doi: 10.1016/S0092-8674(00)80655-8
6. Hollis BW, Wagner CL. Clinical review: the role of the parent compound
vitamin D with respect to metabolism and function: Why clinical dose
intervals can affect clinical outcomes. J Clin Endocrinol Metab. (2013)
98:4619–28. doi: 10.1210/jc.2013-2653
7. Jorde R, Grimnes G. Serum cholecalciferol may be a better marker of
vitamin D status than 25-hydroxyvitamin D. Med Hypotheses (2018) 111:61–5.
doi: 10.1016/j.mehy.2017.12.017
8. Bikle DD, Gee E, Halloran B, Haddad JG. Free 1,25-dihydroxyvitamin D levels
in serum from normal subjects, pregnant subjects, and subjects with liver
disease. J Clin Invest. (1984) 74:1966–71. doi: 10.1172/JCI111617
9. Schwartz JB, Lai J, Lizaola B, Kane L, Weyland P, Terrault NA, et al. Variability
in free 25(OH) vitamin D levels in clinical populations. J Steroid Biochem Mol
Biol. (2014) 144 Pt A:156–8. doi: 10.1016/j.jsbmb.2013.11.006
10. Schmidt-Gayk H, Grawunder C, Tschöpe W, Schmitt W, Ritz E, Pietsch V,
et al. 25-hydroxy-vitamin-D in nephrotic syndrome. Lancet (1977) 2:105–8.
doi: 10.1016/S0140-6736(77)90118-0
11. 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) 96:142–9.
doi: 10.1210/jc.2010-0980
12. Sollid ST, Hutchinson MY, Berg V, Fuskevag OM, Figenschau Y, Thorsby
PM, et al. Effects of vitamin D binding protein phenotypes and vitamin
D supplementation on serum total 25(OH)D and directly measured free
25(OH)D. Eur J Endocrinol. (2016) 174:445–52. doi: 10.1530/EJE-15-1089
13. Mendel CM. The free hormone hypothesis: a physiologically
based mathematical model. Endocr Rev. (1989) 10:232–74.
doi: 10.1210/edrv-10-3-232
14. Cody V. Thyroid hormone interactions: molecular conformation,
protein binding, and hormone action. Endocr Rev. (1980) 1:140–66.
doi: 10.1210/edrv-1-2-140
15. Jones G, Prosser DE, Kaufmann M. 25-Hydroxyvitamin D-24-hydroxylase
(CYP24A1): its important role in the degradation of vitamin D. Arch Biochem
Biophys. (2012) 523:9–18. doi: 10.1016/j.abb.2011.11.003
16. Bikle D, Bouillon R, Thadhani R, Schoenmakers I. Vitamin D metabolites
in captivity? Should we measure free or total 25(OH)D to assess
vitamin D status? J Steroid Biochem Mol Biol. (2017) 173:105–16.
doi: 10.1016/j.jsbmb.2017.01.007
17. Schwartz JB, Lai J, Lizaola B, Kane L, Markova S, Weyland P, et al.
A comparison of measured and calculated free 25(OH) vitamin D
levels in clinical populations. J Clin Endocrinol Metab. (2014) 99:1631–7.
doi: 10.1210/jc.2013-3874
18. Arnaud J, Constans J. Affinity differences for vitamin D metabolites associated
with the genetic isoforms of the human serum carrier protein (DBP). Hum
Genet. (1993) 92:183–8. doi: 10.1007/BF00219689
19. Nielson CM, Jones KS, Chun RF, Jacobs JM, Wang Y, Hewison M, et al.
Free 25-hydroxyvitamin D: impact of vitamin D binding protein assays on
racial-genotypic associations. J Clin Endocrinol Metab. (2016) 101:2226–34.
doi: 10.1210/jc.2016-1104
20. Holick MF. Vitamin D deficiency. N Engl J Med. (2007) 357:266–81.
doi: 10.1056/NEJMra070553
21. Bouillon R, Van Assche FA, Van Baelen H, Heyns W, De Moor P. Influence
of the vitamin D-binding protein on the serum concentration of 1,25-
dihydroxyvitamin D3. Significance of the free 1,25-dihydroxyvitamin D3
concentration. J Clin Invest. (1981) 67:589–96. doi: 10.1172/JCI110072
22. 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)
63:954–9. doi: 10.1210/jcem-63-4-954
23. Powe CE, Ricciardi C, Berg AH, Erdenesanaa D, Collerone G, Ankers E,
et al. Vitamin D-binding protein modifies the vitamin D-bone mineral density
relationship. J Bone Miner Res. (2011) 26:1609–16. doi: 10.1002/jbmr.387
24. Johnsen MS, Grimnes G, Figenschau Y, Torjesen PA, Almas B, Jorde R.
Serum free and bio-available 25-hydroxyvitamin D correlate better with bone
density than serum total 25-hydroxyvitamin D. Scand J Clin Lab Invest. (2014)
74:177–83. doi: 10.3109/00365513.2013.869701
25. Heureux N, Lindhout E, Swinkels L. A direct assay for measuring
free 25-hydroxyvitamin D. J AOAC Int. (2017) 100:1318–22.
doi: 10.5740/jaoacint.17-0084
26. Bland R, Markovic D, Hills CE, Hughes SV, Chan SL, Squires PE,
et al. Expression of 25-hydroxyvitamin D3-1alpha-hydroxylase in
pancreatic islets. J Steroid Biochem Mol Biol. (2004) 89–90:121–5.
doi: 10.1016/j.jsbmb.2004.03.115
27. Johnson JA, Grande JP, Roche PC, Kumar R. Immunohistochemical
localization of the 1,25(OH)2D3 receptor and calbindin D28k in human and
rat pancreas. Am J Physiol. (1994) 267(3 Pt 1):E356–60.
28. Cade C, Norman AW. Vitamin D3 improves impaired glucose tolerance and
insulin secretion in the vitamin D-deficient rat in vivo.Endocrinology (1986)
119:84–90. doi: 10.1210/endo-119-1-84
29. Palomer X, González-Clemente JM, Blanco-Vaca F, Mauricio D.
Role of vitamin D in the pathogenesis of type 2 diabetes mellitus.
Diabetes Obes Metab. (2008) 10:185–97. doi: 10.1111/j.1463-1326.2007.
00710.x
30. Angellotti E, Pittas AG. The role of vitamin D in the prevention of
type 2 diabetes: to D or not to D? Endocrinology (2017) 158:2013–21.
doi: 10.1210/en.2017-00265
31. Afzal S, Brøndum-Jacobsen P, Bojesen SE, Nordestgaard BG.
Vitamin D concentration, obesity, and risk of diabetes: a mendelian
randomisation study. Lancet Diabetes Endocrinol. (2014) 2:298–306.
doi: 10.1016/S2213-8587(13)70200-6
32. Ye Z, Sharp SJ, Burgess S, Scott RA, Imamura F, InterAct Consortium,
et al. Association between circulating 25-hydroxyvitamin D and
incident type 2 diabetes: a mendelian randomisation study. Lancet
Diabetes Endocrinol. (2015) 3:35–42. doi: 10.1016/S2213-8587(14)
70184-6
Frontiers in Endocrinology | www.frontiersin.org 5February 2019 | Volume 10 | Article 79
18
Jorde DBP, Total and Free 25(OH)D and Diabetes
33. Davidson MB, Duran P, Lee ML, Friedman TC. High-dose vitamin D
supplementation in people with prediabetes and hypovitaminosis D. Diabetes
Care (2013) 36:260–6. doi: 10.2337/dc12-1204
34. Jorde R, Sollid ST, Svartberg J, Schirmer H, Joakimsen RM, Njølstad I, et al.
Vitamin D 20,000 IU per week for five years does not prevent progression
from prediabetes to diabetes. J Clin Endocrinol Metab. (2016) 101:1647–55.
doi: 10.1210/jc.2015-4013
35. Lee CC, Young KA, Norris JM, Rotter JI, Liu Y, Lorenzo C, et al. Association
of directly measured plasma free 25(OH)D with insulin sensitivity and
secretion: the IRAS family study. J Clin Endocrinol Metab. (2017) 102:2781–8.
doi: 10.1210/jc.2017-00039
36. Wang TJ, Zhang F, Richards JB, Kestenbaum B, van Meurs JB,
Berry D, et al. Common genetic determinants of vitamin D
insufficiency: a genome-wide association study. Lancet (2010) 376:180–8.
doi: 10.1016/S0140-6736(10)60588-0
37. Pittas AG, Dawson-Hughes B, Sheehan PR, Rosen CJ, Ware JH, Knowler
WC, et al. Rationale and design of the Vitamin D and Type 2 Diabetes
(D2d) study: a diabetes prevention trial. Diabetes Care (2014) 37:3227–34.
doi: 10.2337/dc14-1005
38. Michaëlsson K, Rasmusson A, Wolk A, Byberg L, Mitchell A, Melhus H.
The free hormone hypothesis: is free serum 25-hydroxyvitamin D a better
marker for bone mineral density in older women? JBMR Plus (2018) 2:367–74.
doi: 10.1002/jbm4.10059
39. Best CM, Pressman EK, Queenan RA, Cooper E, O’Brien KO. Longitudinal
changes in serum vitamin D binding protein and free 25-hydroxyvitamin D
in a multiracial cohort of pregnant adolescents. J Steroid Biochem Mol Biol.
(2018)186:79–88. doi: 10.1016/j.jsbmb.2018.09.019.
40. Peris P, Filella X, Monegal A, Guañabens N, Foj L, Bonet M, et al. Comparison
of total, free and bioavailable 25-OH vitamin D determinations to evaluate its
biological activity in healthy adults: the LabOscat study. Osteoporos Int. (2017)
28:2457–64. doi: 10.1007/s00198-017-4062-8
41. Aloia J, Dhaliwal R, Mikhail M, Shieh A, Stolberg A, Ragolia L, et al. Free
25(OH)D and calcium absorption, PTH, and markers of bone turnover. J Clin
Endocrinol Metab. (2015) 100:4140–5. doi: 10.1210/jc.2015-2548
42. Lopez-Molina M, Santillan C, Murillo M, Valls A, Bosch L, Bel J,
et al. Measured free 25-hydroxyvitamin D in healthy children and
relationship to total 25-hydroxyvitamin D, calculated free 25-hydroxyvitamin
D and vitamin D binding protein. Clin Biochem. (2018) 61:23–7.
doi: 10.1016/j.clinbiochem.2018.08.007
43. Shieh A, Ma C, Chun RF, Wittwer-Schegg J, Swinkels L, Huijs T, et al.
Associations between change in total and free 25-hydroxyvitamin D with
24,25-dihydroxyvitamin D and parathyroid hormone. J Clin Endocrinol
Metab. (2018) 103:3368–75. doi: 10.1210/jc.2018-00515
44. Sauer CG, Loop MS, Venkateswaran S, Tangpricha V, Ziegler TR, Dhawan
A, et al. Free and bioavailable 25-hydroxyvitamin D concentrations are
associated with disease activity in pediatric patients with newly diagnosed
treatment naïve ulcerative colitis. Inflamm Bowel Dis. (2018) 24:641–50.
doi: 10.1093/ibd/izx052
45. Eloi M, Horvath DV, Ortega JC, Prado MS, Andrade LE, Szejnfeld VL, et al.
25-hydroxivitamin D serum concentration, not free and bioavailable vitamin
D, is associated with disease activity in systemic lupus erythematosus patients.
PLoS ONE (2017) 12:e0170323. doi: 10.1371/journal.pone.0170323
46. Srikanth P, Chun RF, Hewison M, Adams JS, Bouillon R, Vanderschueren
D, et al. Associations of total and free 25OHD and 1,25(OH)2D with serum
markers of inflammation in older men. Osteoporos Int. (2016) 27:2291–300.
doi: 10.1007/s00198-016-3537-3
47. Yu C, Xue H, Wang L, Chen Q, Chen X, Zhang Y, et al. Serum bioavailable
and free 25-hydroxyvitamin D levels, but not its total level, are associated with
the risk of mortality in patients with coronary artery disease. Circ Res. (2018)
123:996–1007. doi: 10.1161/CIRCRESAHA.118.313558
48. Lee CJ, Iyer G, Liu Y, Kalyani RR, Bamba N, Ligon CB, et al. The effect of
vitamin D supplementation on glucose metabolism in type 2 diabetes mellitus:
a systematic review and meta-analysis of intervention studies. J Diabetes
Complications (2017) 31:1115–26. doi: 10.1016/j.jdiacomp.2017.04.019
49. Williams MH, Van Alstyne EL, Galbraith RM. Evidence of a
novel association of unsaturated fatty acids with Gc (vitamin D-
binding protein). Biochem Biophys Res Commun. (1988) 153:1019–24.
doi: 10.1016/S0006-291X(88)81330-5
50. Van Baelen H, Bouillon R, De Moor P. Vitamin D-binding protein (Gc-
globulin) binds actin. J Biol Chem. (1980) 255:2270–2.
51. Meier U, Gressner O, Lammert F, Gressner AM. Gc-globulin: roles in response
to injury. Clin Chem. (2006) 52:1247–53. doi: 10.1373/clinchem.2005.065680
52. Delanghe JR, Speeckaert R, Speeckaert MM. Behind the scenes of vitamin D
binding protein: more than vitamin D binding. Best Pract Res Clin Endocrinol
Metab. (2015) 29:773–86. doi: 10.1016/j.beem.2015.06.006
53. Nagasawa H, Sasaki H, Uto Y, Kubo S, Hori H. Association of the macrophage
activating factor (MAF) precursor activity with polymorphism in vitamin
D-binding protein. Anticancer Res. (2004) 24:3361–6.
54. Jorde R, Schirmer H, Wilsgaard T, Bøgeberg Mathiesen E, Njølstad I,
Løchen ML, et al. The DBP phenotype Gc-1f/Gc-1f is associated with
reduced risk of cancer. The Tromsø study. PLoS ONE (2015) 10:e0126359.
doi: 10.1371/journal.pone.0126359
55. Wang G, Li Y, Li L, Yu F, Cui L, Ba Y, et al. Association of the
vitamin D binding protein polymorphisms with the risk of type
2 diabetes mellitus: a meta-analysis. BMJ Open (2014) 4:e005617.
doi: 10.1136/bmjopen-2014-005617
56. Leong A, Rehman W, Dastani Z, Greenwood C, Timpson N, Langsetmo L,
et al. The causal effect of vitamin D binding protein (DBP) levels on calcemic
and cardiometabolic diseases: a Mendelian randomization study. PLoS Med.
(2014) 11:e1001751. doi: 10.1371/journal.pmed.1001751
57. Hewison M, Gacad MA, Lemire J, Adams JS. Vitamin D as a cytokine
and hematopoetic factor. Rev Endocr Metab Disord. (2001) 2:217–27.
doi: 10.1023/A:1010015013211
58. Altieri B, Muscogiuri G, Barrea L, Mathieu C, Vallone CV, Mascitelli L,
et al. Does vitamin D play a role in autoimmune endocrine disorders?
A proof of concept. Rev Endocr Metab Disord. (2017) 18:335–46.
doi: 10.1007/s11154-016-9405-9
59. Thorsen SU, Mårild K, Olsen SF, Holst KK, Tapia G, Granström C, et al.
Lack of association between maternal or neonatal vitamin d status and risk of
childhood type 1 diabetes: a scandinavian case-cohort study. Am J Epidemiol.
(2018) 187:1174–81. doi: 10.1093/aje/kwx361
60. Jacobsen R, Thorsen SU, Cohen AS, Lundqvist M, Frederiksen P, Pipper CB,
et al. Neonatal vitamin D status is not associated with later risk of type 1
diabetes: results from two large Danish population-based studies. Diabetologia
(2016) 59:1871–81. doi: 10.1007/s00125-016-4002-8
61. Hyppönen E, Läärä E, Reunanen A, Järvelin MR, Virtanen SM. Intake of
vitamin D and risk of type 1 diabetes: a birth-cohort study. Lancet (2001)
358:1500–3. doi: 10.1016/S0140-6736(01)06580-1
62. Dong JY, Zhang WG, Chen JJ, Zhang ZL, Han SF, Qin LQ. Vitamin D intake
and risk of type 1 diabetes: a meta-analysis of observational studies. Nutrients
(2013) 5:3551–62. doi: 10.3390/nu5093551
63. Littorin B, Blom P, Schölin A, Arnqvist HJ, Blohmé G, Bolinder J, et al. Lower
levels of plasma 25-hydroxyvitamin D among young adults at diagnosis of
autoimmune type 1 diabetes compared with control subjects: results from the
nationwide Diabetes Incidence Study in Sweden (DISS). Diabetologia (2006)
49:2847–52. doi: 10.1007/s00125-006-0426-x
64. Shen L, Zhuang QS, Ji HF. Assessment of vitamin D levels in type 1 and
type 2 diabetes patients: results from metaanalysis. Mol Nutr Food Res. (2016)
60:1059–67. doi: 10.1002/mnfr.201500937
65. Gregoriou E, Mamais I, Tzanetakou I, Lavranos G, Chrysostomou S. The
effects of vitamin D supplementation in newly diagnosed type 1 diabetes
patients: systematic review of randomized controlled trials. Rev Diabet Stud.
(2017) 14:260–8. doi: 10.1900/RDS.2017.14.260
66. Penna-Martinez M, Badenhoop K. Inherited variation in vitamin
D genes and type 1 diabetes predisposition. Genes (2017) 8:E125.
doi: 10.3390/genes8040125
67. Cooper JD, Smyth DJ, Walker NM, Stevens H, Burren OS, Wallace C, et al.
Inherited variation in vitamin D genes is associated with predisposition
to autoimmune disease type 1 diabetes. Diabetes (2011) 60:1624–31.
doi: 10.2337/db10-1656
68. Ongagna JC, Pinget M, Belcourt A. Vitamin D-binding protein gene
polymorphism association with IA-2 autoantibodies in type 1 diabetes.
Clin Biochem. (2005) 38:415–9. doi: 10.1016/j.clinbiochem.2004.
12.013
69. Ongagna JC, Kaltenbacher MC, Sapin R, Pinget M, Belcourt A. The
HLA-DQB alleles and amino acid variants of the vitamin D-binding
Frontiers in Endocrinology | www.frontiersin.org 6February 2019 | Volume 10 | Article 79
19
Jorde DBP, Total and Free 25(OH)D and Diabetes
protein in diabetic patients in Alsace. Clin Biochem. (2001) 34:59–63.
doi: 10.1016/S0009-9120(00)00197-1
70. Blanton D, Han Z, Bierschenk L, Linga-Reddy MV, Wang H, Clare-
Salzler M, et al. Reduced serum vitamin D-binding protein levels are
associated with type 1 diabetes. Diabetes (2011) 60:2566–70. doi: 10.2337/
db11-0576
71. Verhaeghe J, van Herck E, Visser WJ, Suiker AM, Thomasset M, Einhorn
TA, et al. Bone and mineral metabolism in BB rats with long-term diabetes.
Decreased bone turnover and osteoporosis. Diabetes (1990) 39:477–82.
doi: 10.2337/diab.39.4.477
72. Jorde R. RCTS are the only appropriate way to demonstrate the role
of vitamin D in health. J Steroid Biochem Mol Biol. (2018) 177:10–14.
doi: 10.1016/j.jsbmb.2017.05.004
73. Premer C, Schulman IH. Have we been measuring the wrong form of vitamin
D? Circ Res. (2018) 123:934–5. doi: 10.1161/CIRCRESAHA.118.313814
Conflict of Interest Statement: The author declares that the research was
conducted in the absence of any commercial or financial relationships that could
be construed as a potential conflict of interest.
Copyright © 2019 Jorde. This is an open-access article distributed under the terms
of the Creative Commons Attribution License (CC BY). The use, distribution or
reproduction in other forums is permitted, provided the original author(s) and the
copyright owner(s) are credited and that the original publication in this journal
is cited, in accordance with accepted academic practice. No use, distribution or
reproduction is permitted which does not comply with these terms.
Frontiers in Endocrinology | www.frontiersin.org 7February 2019 | Volume 10 | Article 79
20
ORIGINAL RESEARCH
published: 27 March 2019
doi: 10.3389/fendo.2019.00171
Frontiers in Endocrinology | www.frontiersin.org 1March 2019 | Volume 10 | Article 171
Edited by:
Jonathan H. Tobias,
University of Bristol, United Kingdom
Reviewed by:
Jan Josef Stepan,
Charles University, Czechia
Michaël R. Laurent,
University Hospitals Leuven, Belgium
*Correspondence:
Xiangbing Wang
wangx9@rwjms.rutgers.edu
Specialty section:
This article was submitted to
Bone Research,
a section of the journal
Frontiers in Endocrinology
Received: 07 January 2019
Accepted: 01 March 2019
Published: 27 March 2019
Citation:
Wang X, Sheng Z, Meng L, Su C,
Trooskin S and Shapses SA (2019)
25-Hydroxyvitamin D and Vitamin D
Binding Protein Levels in Patients With
Primary Hyperparathyroidism Before
and After Parathyroidectomy.
Front. Endocrinol. 10:171.
doi: 10.3389/fendo.2019.00171
25-Hydroxyvitamin D and Vitamin D
Binding Protein Levels in Patients
With Primary Hyperparathyroidism
Before and After Parathyroidectomy
Xiangbing Wang 1
*, Zhifeng Sheng 2, Lingqiong Meng 3, Chi Su 1, Stanley Trooskin4and
Sue A. Shapses 3
1Divisions of Endocrinology, Metabolism, and Nutrition, Departments of Medicine and Surgery, Rutgers-Robert Wood
Johnson Medical School, New Brunswick, NJ, United States, 2Institution of Metabolism and Endocrinology, The Second
Xiangya Hospital, Central South University, Changsha, China, 3Department of Nutritional Sciences, Rutgers University, New
Brunswick, NJ, United States, 4Divisions of General Surgery, Departments of Medicine and Surgery, Rutgers-Robert Wood
Johnson Medical School, New Brunswick, NJ, United States
Objective: To evaluate vitamin D binding protein and free 25-hydroxyvitamin D [25(OH)D]
levels in healthy controls compared to primary hyperparathyroidism (PHPT) patients, and
to examine PHPT before and after surgery.
Methods: Seventy-five PHPT patients and 75 healthy age, gender, and body mass
index (BMI) -matched control subjects were examined. In addition, 25 PHPT patients
underwent parathyroidectomy and had a 3-month follow up visit. Levels of total and free
25(OH)D, DBP, and intact parathyroid hormone (iPTH) were determined before and 3
months after surgery.
Results: There was no significant difference in age and BMI between PHPT patients and
controls. Levels of 25(OH)D and DBP were lower in PHPT patients compared to controls
(p<0.01). There was no significant difference in calculated free and bioavailable 25(OH)D
levels between PHPT patients and controls. Calcium and iPTH levels decreased to normal
but DBP and DBP-bound-25(OH)D increased (P<0.001) after parathyroidectomy.
Levels of DBP were inversely correlated with iPTH (r= −0.406, P<0.001) and calcium
levels (r= −0.423, P<0.001).
Conclusion: Serum DBP levels were lower in patients with PHPT and
parathyroidectomy restored DBP levels. We suggest that lower DBP levels is one
of contributing mechanisms of low total 25(OH)D in PTHP patients and the total
25(OH)D levels might not reflect true vitamin D status in PHPT patients.
Keywords: vitamin D binding protein, vitamin D deficiency, parathyroid hormone, calcium metabolism,
hyperparathyroidism, parathyroidectomy
INTRODUCTION
Total 25(OH)D level has been recognized as an optimal indicator of vitamin D nutrition status,
and lower 25(OH)D concentration is usually considered as vitamin D deficiency or insufficiency
in clinical practice. Low total 25(OH)D concentration, which is common in PHPT patients, is
associated with the severity of the disease and high parathyroid adenoma weight (13). Low
21
Wang et al. DBP and 25OHD in PHPT Patients
25(OH)D levels also exist in many chronic conditions such as
end-stage liver disease and nephrotic syndrome, and in critical
illness where intact parathyroid hormone levels are not elevated
(4,5). The majority of circulating 25(OH)D tightly bound to
DBP, with a smaller amount (10–15%) bound to albumin. Less
than 1% of circulating vitamin D metabolites exists in a free,
unbound form (5). The variations in the 25(OH)D levels in these
conditions result from variations in the binding of 25(OH)D to
DBP (4).
Previous studies showed that DBP are lower in PHPT patients
compared with age, BMI, and gender matched (68) or genetic
background matched controlled subjects (7). It is unclear how
DBP is regulated in and if the elevated iPTH plays a role in PHPT,
or if DBP simply a biomarker of circulating 25(OH)D. Since the
majority (>85%) of circulating 25(OH)D is bound to DBP, we
suggest that decreased DBP might be one of mechanisms of low
total 25(OH)D levels in PHPT patient (8). The causes of lower
DBP concentration in the serum of PHPT patients remained
unknown. We hypothesize that the elevated iPTH or calcium
levels inhibit DBP production in the liver of PHPT patient. In
our current study, we investigated the effects of lower calcium
and iPTH levels by parathyroidectomy on DPB in PHPT patients.
We also compared levels of 25(OH)D, DBP, and calculated free
and bioavailable 25(OH)D in patients with PHPT with normal
controls. The aim of this study is to investigate the effects of
parathyroidectomy on DBP and DBP-bound 25(OH)D levels in
PHPT patients.
METHODS
Study Subjects
Seventy-five PHPT patients (61 Caucasians, eight African
American, four Asians, and two Hispanic Americans) were
seen at the Endocrinology and General Surgery clinics of
Robert Wood Johnson University Hospital from January 2010 to
December 2016 at prior to treatment. Most of the patients were
relatively asymptomatic with less severe PHPT profile (9). The
inclusion criteria were: (1) a serum calcium level >10.6 mg/dL
(8.6–10.4 mg/dl) and intact PTH (iPTH) >66 pg/mL (15–65
pg/mL), (2) age 20–80 years, and (3) 24-h urine calcium >100 mg
(100–300 mg/24 h) with fraction excretion of calcium >0.01.
The exclusion criteria were: (1) hormone replacement therapy
or contraceptive pills, (2) hepatic dysfunction, or (3) renal
dysfunction, and (4) BMI >40 (kg/m2). Seventy-five age, gender,
and BMI-matched healthy volunteers (62 Caucasians, eight
African Americans, four Asians, and one Hispanic American)
(10) from the community were included as controls after a
multistep screening process and did not take contraceptive
pills. The healthy controls took 400 IU vitamin D supplement.
Supplemental vitamin D intake in patients before surgery is
not known. Twenty-five PHPT patients (seven males and 18
females) underwent parathyroidectomy (PTX) monitored by
intro-operative iPHT levels and were examined at 3 months
during their follow-up visit after surgery. All minimally invasive
PTX were done by one surgeon and all patients were advised to
take 0.25 mcg calcitriol for 1–2 weeks and 1,000–2,000 IU vitamin
D for 1–3 months after PTX to prevent hypocalcemia as standard
post-operative clinical care (11). All subjects and patients signed
an informed consent and the use of human subjects in this study
was approved by the IRB at Rutgers University.
Sample Collections and Assays
Venous blood samples were collected from patients and
controlled subjects after a 12-h overnight fast. Twenty-five PHPT
patients had parathyroid surgery and finished 3 months’ post-
surgery follow up visit. Serum was separated and stored at 70 C
for measurement of 25(OH)D and DBP levels. Intact-PTH,
serum calcium, and albumin were determined by commercial
laboratories. The laboratory uses both internal and external
standards, and also participated in the international Vitamin
D External Quality Assessment Scheme to ensure the quality
and accuracy of the 25(OH)D analysis and serum 25(OH)D
levels (radioimmunoassay; DiaSorin) CV <12.5%). DBP levels
in serum were determined using a commercial polyclonal ELISA
kit (ALPCO, Salem, NH). The intra- and inter-assay coefficients
of variation are 5.0 and 12.7%, respectively. Free, bioavailable,
albumin-bound and DBP-bound 25(OH)D concentrations were
calculated using equations adapted from Bikle et al. (12).
Statistical Analyses
Results are expressed as mean ±SD. Shapiro-Wilk test was used
to check normality. Two-tailed Student’s t-test and Wilcoxon
Rank Sum test were used to compare values between groups with
normally and non-normally distribution, respectively. Changes
before and after parathyroidectomy were compared with a paired
Student’s t-test. Correlation coefficients and linear regression
were used to assess relationships between variables. A P<0.05
was defined as the level of significance. Statistical analysis was
performed with SAS v9.4.
TABLE 1 | Subject characteristics and serum concentrations.
Variable Control PHPT P-value
n=75 n=75
Age (years) 58.0 ±8.1 59.3 ±12.3 0.314
BMI (kg/m2) 29.9 ±2.1 30.6 ±4.8 0.373
Calcium (mg/dL) 9.4 ±0.5 11.1 ±0.6 <0.001
iPTH (pg/mL) 37.9 ±17.3 140.4 ±70.5 <0.001
25(OH)D (ng/mL) 28.3 ±5.4 23.6 ±8.3 <0.001
DBP (mg/dL) 42.1 ±7.0 35.2 ±7.9 <0.001
Albumin (g/dL) 4.5 ±0.2 4.3 ±0.3 <0.001
DBP-bound
25(OH)D (ng/mL)
26.4 ±5.1 21.8 ±7.7 <0.001
Albumin-bound
25(OH)D (ng/mL)
1.9 ±0.5 1.8 ±0.8 0.082
Bioavailable
25(OH)D (ng/mL)
1.9 ±0.5 1.8 ±0.8 0.083
Free 25(OH)D
(pg/mL)
4.7 ±1.1 4.7 ±2.0 0.573
BMI, body mass index; iPTH, intact parathyroid hormone; 25(OH)D, 25-hydroxyvitamin
D; DBP
, vitamin D binding protein. Data shows as mean ±standard deviation.
Frontiers in Endocrinology | www.frontiersin.org 2March 2019 | Volume 10 | Article 171
22
Wang et al. DBP and 25OHD in PHPT Patients
RESULTS
Seventy-five PHPT patient (23 men, 11 premenopausal women,
and 41 postmenopausal women) and 75 control subjects (19 men,
11 premenopausal women, and 45 postmenopausal women) were
included in this study. The mean concentrations of calcium,
albumin, 25(OH)D, iPTH, and DBP determined in the serum
samples from control subjects and PHPT patients are shown
in (Table 1). Both total 25(OH)D and DBP levels were about
17% lower in PHPT patients compared to control subjects
(P<0.001). There was no significant difference in albumin-
bound 25(OH)D, but DBP-bound 25(OH)D was also 17%
lower in PTHP patients compared to control subjects (P<
0.001). In addition, albumin levels were significantly lower in
PHPT patients than in control subjects (p<0.001). There
were no significant differences between bioavailable and free
25(OH)D between healthy controls and PHPT patients (Table 1).
Comparison of individual 25(OH), DBP, free 25(OH)D, and
bioavailable 25(OH) D between healthy controls and PHPT
patients were showed in Figure 1.
FIGURE 1 | (A-D) Comparison of serum levels of DBP and 25 (OH) D between PHPT patients and control subjects. DBP, vitamin D binding protein; 25OHD,
25-hydroxyvitamin D. ***P<0.001.
TABLE 2 | Spearman correlation coefficients between DBP and other variables (n=150).
Age BMI Calcium iPTH 25(OH)D Albumin Free 25(OH)D Bioavailable
25(OH)D
DBP 0.210a0.192a0.423c0.406c0.253b0.139a0.344c0.295c
Age 0.113 0.076 0.055 0.132 0.074 0.301c0.278c
BMI 0.038 0.074 0.272c0.141 0.121 0.145
Calcium 0.751c0.292c0.218b0.057 0.114
iPTH 0.418c0.312c0.153 0.233b
25(OH)D 0.225b0.767c0.786c
Albumin 0.092 0.284c
Free
25(OH)D
0.972c
BMI, body mass index; iPTH, intact parathyroid hormone; 25(OH)D, 25-hydroxyvitamin D; DBP, vitamin D binding protein. Bold means significan. aP<0.05; bP<0.01; cP<0.001.
Frontiers in Endocrinology | www.frontiersin.org 3March 2019 | Volume 10 | Article 171
23
Wang et al. DBP and 25OHD in PHPT Patients
Levels of DPB (n=150) were positively correlated with total
25(OH)D (r=0.253, P<0.01) but inversely correlated with
iPTH (r= −0.406, P<0.001) and calcium (r= −0.423, P<
0.001; Table 2). Levels of iPTH inversely correlated with total
25(OH)D (r= −0.418, P<0.001) and bioavailable 25(OH)D (r
= −0.233, P<0.01; Table 2).
In PHPT patients who underwent parathyroidectomy, serum
iPTH, and calcium decreased to normal but DBP levels increased
by 15% (P<0.01, Table 3). Serum total 25(OH) D were increased
by 43 % (p<0.001) but DBP-bound 25(OH)D also increased
by 43% (P=0.001). As a result, there was an attenuated rise
in bioavailable (23%, P=0.024) and free 25(OH)D (21%, P
=0.021, Table 3). Comparison of individual 25(OH), DBP, free
25(OH)D, and bioavailable 25(OH) D before and after PTX was
showed in Figure 2. Multiple regression showed that none of
the variables (Ca, PTH or 25(OH)D) predicted the change in
DBP after parathyroidectomy (not shown). In addition, there
were no predictors for the rise in 25(OH)D due to surgery.
Multiple Regression indicated that only the change in albumin
predicted change in bioavailable 25(OH)D (p=0.027), but not
free 25(OH)D (p=0.122) after parathyroidectomy.
DISCUSSION
The results of our current study demonstrate that PHPT patients
have lower serum levels of DBP and total serum 25(OH)D
consistent with our previous study (8) and study by Battista et al.
(7). In addition, our data confirmed our previous studies that
the calculated free or bioavailable 25(OH)D remained unchanged
compared with normal control subjects (8). We also showed
that PTX increases DBP and DBP-bound 25(OH)D levels. Thus,
based on our findings and because supplemental vitamin D raises
25(OH)D, but not DBP (1315), we suggest that the increased
DBP level after PTX might be due to the decreased iPTH or
lowered calcium levels after surgery. This supports the hypothesis
that DBP is not simply a biomarker of 25(OH)D. Our current
TABLE 3 | PHPT profile changes before and after parathyroidectomy.
Before After P-value
n=25 n=25
BMI (kg/ m2) 31.0 ±5.6 29.0 ±4.3 0.127
Calcium (mg/ dl) 11.0 ±0.6 9.7 ±0.4 <0.001
iPTH (pg/ ml) 121.5 ±40.5 44.7 ±8.2 <0.001
25(OH)D (ng/ ml) 26.4 ±7.8 37.7 ±12.1 <0.001
DBP (mg/ dl) 38.9 ±9.8 44.7 ±8.2 0.001
Albumin (g/ dl) 4.3 ±0.9 4.5 ±0.2 0.046
DBP-bound 25(OH)D
(ng/ mL)
24.6 ±7.3 35.4 ±11.1 <0.001
Albumin-bound
25(OH)D (ng/ mL)
2.0 ±0.5 2.36 ±0.75 0.024
Bioavailable 25(OH)D
(ng/mL)
1.94 ±0.94 2.36 ±0.74 0.024
Free25(OH)D (pg/mL) 4.89 ±2.05 5.90 ±1.9 0.021
BMI, body mass index; DBP, vitamin D binding protein; iPTH, intact parathyroid hormone;
25(OH)D, 25-hydroxyvitamin D. Data are means ±standard deviation (Paired t-test).
results also support the concept that the lower DBP levels in
PHPT compared to healthy matched controls may be one of
the factors contributing to the low total 25(OH)D levels in
PHPT patients. Another possible factor leading to the low total
25(OH)D levels in PHPT patients include the conversion to 1,25
or 24,25 (OH)2D due to elevated iPTH or FGF-23 (6,16).
Aloia et al. found that black Americans have lower levels of
total 25(OH)D but the free 25(OH)D remains relative unchanged
by direct measurement of free 25(OH)D (17). Pre-menopausal
women have higher serum DBP, estradiol, and 25(OH)D levels
than postmenopausal women (18). The calculated free 25OHD
was also lower in postmenopausal women than that of control
subjects, but to a much lesser degree than total 25OHD (18).
In a recent study, Pilz et al. found that women taking estrogen
containing contraceptive measures have higher total 25(OH)D
but unaltered free 25(OH)D levels by direct measurement (19).
The results suggest that total 25(OH)D levels might not be
an accurate marker of bioactive vitamin D status in at least
a few situations, including black Americans or women taking
hormonal contraceptive pill or other clinical situations (5,20)
Bioavailable 25(OH)D may be a better measure of vitamin D
status with respect to bone mineral density (BMD) and mineral
metabolism, as has been shown in nephrotic syndrome patients
(21). Lai et al. found that cirrhosis patients with low albumin had
lower DBP, total 25(OH)D, and free 25(OH)D levels and suggest
that total 25(OH)D is not accurate marker for vitamin D status
in these patients (22). Yu et al. reported that it is bioavailable
and free 25(OH)D levels, not total 25(OH), associated with the
risk of mortality in Chinese patients with coronary artery disease
(23). Our results suggest that the total 25(OH)D levels in PHPT
patients may not be a good indicator of vitamin D status before or
after surgery since there is a much lower rise in both bioavailable
and free 25(OH)D concentrations.
The appropriate management of asymptomatic PHPT still
require more evidence from clinical studies (1,24) despite the
guidelines for PHPT have been revised recently (25). There are
controversies about vitamin D supplementation in the PTHP
patient with low 25(OH) levels. Marcocci et al. reviewed three
studies; two demonstrated that vitamin D supplementation had
no significant influence on serum and urinary calcium levels, and
one study showed no clinical benefit, while in the third study of
27 PHPT patients, 12 patients developed either increased serum
calcium levels or increased urine calcium excretion (24). Our data
show here that there are no significant differences in free and
bioavailable 25(OH)D levels between PHPT patients and control
subjects. Given the pre-existing high serum Ca in PHPT patients,
we suggest that clinicians should be aware of this treating PHPT
patients with vitamin D, especially when using a loading dose of
vitamin D supplementation (26).
The DBP concentration is relatively stable throughout life
but is altered by gender, menopausal status (10), and genetic
backgrounds (2729). In the current study, we matched PHPT
patients and control subjects for these factors. The underlying
mechanism for lower DBP concentration in PHPT patients,
at least in part, may be explained by the higher iPTH levels
inhibition of liver-derived DBP in PHPT patient, a finding
that has been reported previously (8) and PTH/PTH-related
Frontiers in Endocrinology | www.frontiersin.org 4March 2019 | Volume 10 | Article 171
24
Wang et al. DBP and 25OHD in PHPT Patients
FIGURE 2 | (A-D) Comparison of serum levels of DBP and 25(OH)D before and after parathyroidectomy. DBP, vitamin D binding protein; 25OHD, 25-hydroxyvitamin
D. *P<0.05; **P<0.01; ***P<0.001.
protein receptor is highly expressed in the liver (30). Conditions
such as malnutrition and liver failure might affect DBP,
albumin, and other liver-specific protein status (22). Serum DBP
concentrations were inversely correlated with iPTH and calcium
levels and DBP increased after decreasing iPTH and calcium
by parathyroid surgery. It is also possible that a reduced DPB
is a compensatory mechanism in PHPT to ensure that under
conditions of low total 25(OH)D, there is adequate circulating
free or bioavailable 25(OH)D. Also, the mechanism regulating
the rise in DBP after parathyroidectomy remains unclear, but
it is suggested that studying this population may help to better
understand the binding protein and its regulation of normal
vitamin D metabolism.
The limitations of the study are the relatively small sample
size, including only 25 PHPT patients had 3 months’ post-
surgery data. Total 25OHD levels were measured by RIA, and
not by mass spectrometry which is considered more accurate,
but we used internal and external controls to increase accuracy.
Another limitation is that this study does not include serum
phosphate or FGF-23 levels and the study design cannot confirm
a mechanism of low total 25(OH)D or DBP in PHPT. All
patients took calcitriol for 1–2 weeks after surgery and advised
to take a vitamin D supplement, as standard post-operative
clinical care (11) to prevent risk of low serum calcium levels.
As a result, this may be another reason for the increase in
serum total 25(OH)D at 3 months after parathyroidectomy.
Moreover, calculated free 25(OH)D utilize equations that use
average binding coefficients for DBP and albumin may not be as
accurate as direct measurements (27).
In conclusion, total 25(OH)D and DBP levels are lower in
PHPT patients but calculated free 25(OH) remained relatively
unchanged. Parathyroidectomy increased DBP and DBP-bound
25(OH)D levels. Further research is required to investigate
whether free 25OHD is the better marker of vitamin D status in
the PHPT patient.
AUTHOR CONTRIBUTIONS
ZS, LM, and CS contributed to recruiting patients, data collection
and analysis, and manuscript preparation. ST contributed to
Parathyroidectomy and manuscript preparation. SS contributed
to experimental design, recruiting control subjects, data analysis
and manuscript preparation. XW contributed to experimental
design, data analysis and manuscript preparation.
ACKNOWLEDGMENTS
We thank Dr. Wei Sun MD., Ph.D. for assisted in DBP ELISA
assay; Aseel Al-Dayyeni MD, and Aliza Rubin, RN for help
recruiting PHPT patients, Professor Huizgou Fan for assisted in
the preparation of this manuscript.
Frontiers in Endocrinology | www.frontiersin.org 5March 2019 | Volume 10 | Article 171
25
Wang et al. DBP and 25OHD in PHPT Patients
REFERENCES
1. Bilezikian JP. Primary hyperparathyroidism. J Clin Endocrinol Metab. (2018)
103:3993–4004. doi: 10.1210/jc.2018-01225
2. Rao DS, Honasoge M, Divine GW, Phillips ER, Lee MW, Ansari MR,
et al. Effect of vitamin D nutrition on parathyroid adenoma weight:
pathogenetic and clinical implications. J Clin Endocrinol Metab. (2000)
85:1054–8. doi: 10.1210/jc.85.3.1054
3. Eastell R, Brandi ML, Costa AG, D’Amour P, Shoback DM, Thakker RV.
Diagnosis of asymptomatic primary hyperparathyroidism: proceedings of the
Fourth International Workshop. J Clin Endocrinol Metab. (2014) 99:3570–9.
doi: 10.1210/jc.2014-1414
4. Youselzadeh P, Shapses SA, Wang X. Vitamin D binding protein impact
on 25-hydroxyvitamin D levels under different physiologic and pathologic
conditions. Int J Endocrinol. (2014) 2014:981581.doi: 10.1155/2014/981581
5. Jassil N, Sharma A, Bikle D, Wang X. Vitamin D binding protein and 25-
hydroxyvitamin D levels: emerging clinical applications. Endocr Pract. (2017)
23:605–61. doi: 10.4158/EP161604.RA
6. Wang X, Shapses SA, Wei S, Sukumar D, Ghosh J. Vitamin D binding protein
levels in female patients with primary hyperparathyroidism. Endocr Pract.
(2013) 19:609–13. doi: 10.4158/EP12371.OR
7. Battista C, Guarnieri V, Carnevale V, Baorda F, Pileri M, Garrubba
M, et al. Vitamin D status in primary hyperparathyroidism: effect of
genetic background. Endocrine. (2017) 55:266–72. doi: 10.1007/s12020-016-
0974-x
8. Wang X, Shapses SA, Alhraishawi H. Free and bioavailable 25-
hydroxyvitamin D Levels in patients with primary hyperparathyroidism.
Endocr Pract. (2017) 23:66–71. doi: 10.4158/EP161434.OR
9. Meng L, Al-Dayyeni A, Sheng Z, Wang X. Comparison of initial
clinical presentations between primary hyperparathyroidism patients
from new brunswick and changsha. Int J Endocrinol. (2018)
2018:628687.doi: 10.1155/2018/628687
10. Bolland MJ, Grey AB, Ames RW, Horne AM, MasonBH, Wattie DJ, et al. Age-
, gender-, and weight-related effects on concentrations of 25-hydroxyvitamin
D are not mediated by vitamin D binding protein. Clin Endocrinol. (2007)
67:259–64. doi: 10.1111/j.1365-2265.2007.02873.x
11. Wilhelm SM, Wang TS, Ruan DT, Lee JA, Asa SL, Duh QY, et al.
The american association of endocrine surgeons guidelines for definitive
management of primary hyperparathyroidism. JAMA Surg. (2016) 151:959–
68. doi: 10.1001/jamasurg.2016.2310
12. 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)
63:954–9. doi: 10.1210/jcem-63-4-954
13. Nielson CM, Jones KS, Chun RF, Jacobs JM, Wang Y, Hewison M, et al.
Free 25-hydroxyvitamin D: impact of vitamin D binding protein assays on
racial-genotypic associations. J Clin Endocrinol Metab. (2016) 101:2226–34.
doi: 10.1210/jc.2016-1104
14. Björkhem-Bergman L, Torefalk E, Ekström L, Bergman P. Vitamin D binding
protein is not affected by high-dose vitamin D supplementation: a post hoc
analysis of a randomised, placebo-controlled study. BMC Res Notes. (2018)
11:619. doi: 10.1186/s13104–018-3725–7
15. Pop C, Sukumar D, Schneder SH, Schlussel Y, Stahl T, Gordon C, et al. Three
doses of vitamin D (3DD) on bone mineral density (BMD) and geometry in
older women during modest weight loss. Osteoporosis Int. (2017) 28: 377–88.
doi: 10.1007/s00198-016-3735-z
16. Witteveen JE, van Lierop AH, Papapoulos SE, Hamdy NA.
Increased circulating levels of FGF23: an adaptive response in
primary hyperparathyroidism? Eur J Endocrinol. (2012) 166:55–60.
doi: 10.1530/EJE-11-0523
17. Aloia J, Mikhail M, Dhaliwal R, Shieh A, Usera G, Stolberg A, et al. Free
25(OH)D and the vitamin D paradox in african americans. J Clin Endocrinol
Metab. (2015) 100:3356–63. doi: 10.1210/JC.2015-2066
18. Pop LC, Shapses SA, Chang B, Sun W, Wang X. Vitamin D binding protein
in healthy pre- and postmenopausal women: relationship with estradiol
concentrations. Endocr Pract. (2015) 21:936–42. doi: 10.4158/EP15623.OR
19. Pilz SF, Obeid R, Schwetz V, Trummer C, Pandis ML, Lerchbaum EB, et al
Hormonal contraceptive use is associated with higher total but unaltered free
25-hydroxyvitamin D serum concentrations. J Clin Endocrinol Metab. (2018)
103:2385–91. doi: 10.1210/jc.2018-00336
20. Schwartz JB, Gallagher C, Jorde R, Berg V, Walsh J, Eastell 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)
103:3278–88. doi: 10.1210/jc.2018–00295
21. Aggarwal A, Yadav AK, Ramachandran R, Kumar V, Kumar V, Sachdeva
N, et al. Bioavailable vitamin D levels are reduced and correlate with bone
mineral density and markers of mineral metabolism in adults with nephrotic
syndrome. Nephrology. (2016) 21:483–9. doi: 10.1111/nep.12638
22. Lai JC, Bikle DD, Lizaola B, Hayssen H, Terrault NA, Schwartz JB. Total
25(OH) vitamin D, free 25(OH) vitamin D and markers of bone turnover in
cirrhotics with and without synthetic dysfunction. Liver Int. (2015) 35:2294–
300. doi: 10.1111/liv.12819
23. Yu C, Xue H, Wang L, Chen Q, Chen X, Zhang Y, et al. Serum bioavailable
and free 25-hydroxyvitamin D levels, but not its total level, are associated with
the risk of mortality in patients with coronary artery disease. Circ Res. (2018)
123:996–1007. doi: 10.1161/CIRCRESAHA.118.313558
24. Marcocci C, Bollerslev J, Khan AA, Shoback DM. Medical management
of primary hyperparathyroidism: proceedings of the fourth
International Workshop on the Management of Asymptomatic Primary
Hyperparathyroidism. J Clin Endocrinol Metab. (2014) 99:3607–18.
doi: 10.1210/jc.2014-1417
25. Bilezikian JP, Brandi ML, Eastell R, Silverberg SJ, Udelsman R,
Marcocci C, et al. Guidelines for the management of asymptomatic
primary hyperparathyroidism: summary statement from the Fourth
International Workshop. J Clin Endocrinol Metab. (2014) 99:3561–9.
doi: 10.1210/jc.2014-1413
26. Bischoff-Ferrari HA, Dawson-Hughes B, Orav EJ, Staehelin HB, Meyer OW,
Theiler R, et al. Monthly high-dose vitamin d treatment for the prevention
of functional decline: a randomized clinical trial. JAMA Intern Med. (2016)
176:175–83. doi: 10.1001/jamainternmed.2015.7148
27. Bikle D, Bouillon R, Thadhani R, Schoenmakers I. Vitamin D metabolites
in captivity? Should we measure free or total 25(OH)D to assess
vitamin D status? J Steroid Biochem Mol Biol. (2017) 173:105–16.
doi: 10.1016/j.jsbmb.2017.01.007
28. Schwartz JB, Lai J, Lizaola B, Kane L, Markova S, Weyland P, et al.
A comparison of measured and calculated free 25(OH) vitamin D
levels in clinical populations. J Clin Endocrinol Metab. (2014) 99:1631–7.
doi: 10.1210/jc.2013-3874
29. Bhan I. Vitamin d binding protein and bone health. Int J Endocrinol. (2014)
2014:561214. doi: 10.1155/2014/561214
30. Mitnick MA, Grey A, Masiukiewicz U, Bartkiewicz M, Rios-Velez L, Friedman
S, et al. Parathyroid hormone induces hepatic production of bioactive
interleukin-6 and its soluble receptor. Am J Physiol Endocrinol Metab. (2001)
280:E405–12. doi: 10.1152/ajpendo.2001.280.3.E405
Conflict of Interest Statement: The authors declare that the research was
conducted in the absence of any commercial or financial relationships that could
be construed as a potential conflict of interest.
Copyright © 2019 Wang, Sheng, Meng, Su, Trooskin and Shapses. This is an open-
access article distributed under the terms of the Creative Commons Attribution
License (CC BY). The use, distribution or reproduction in other forums is permitted,
provided the original author(s) and the copyright owner(s) are credited and that the
original publication in this journal is cited, in accordance with accepted academic
practice. No use, distribution or reproduction is permitted which does not comply
with these terms.
Frontiers in Endocrinology | www.frontiersin.org 6March 2019 | Volume 10 | Article 171
26
REVIEW
published: 28 May 2019
doi: 10.3389/fendo.2019.00317
Frontiers in Endocrinology | www.frontiersin.org 1May 2019 | Volume 10 | Article 317
Edited by:
Giacomina Brunetti,
University of Bari Aldo Moro, Italy
Reviewed by:
Ian R. Reid,
The University of Auckland,
New Zealand
Jan Josef Stepan,
Charles University, Czechia
*Correspondence:
Daniel David Bikle
daniel.bikle@ucsf.edu
Specialty section:
This article was submitted to
Bone Research,
a section of the journal
Frontiers in Endocrinology
Received: 25 March 2019
Accepted: 01 May 2019
Published: 28 May 2019
Citation:
Bikle DD and Schwartz J (2019)
Vitamin D Binding Protein, Total and
Free Vitamin D Levels in Different
Physiological and Pathophysiological
Conditions. Front. Endocrinol. 10:317.
doi: 10.3389/fendo.2019.00317
Vitamin D Binding Protein, Total and
Free Vitamin D Levels in Different
Physiological and Pathophysiological
Conditions
Daniel David Bikle 1,2
*and Janice Schwartz 1
1Department of Medicine, University of California, San Francisco, San Francisco, CA, United States, 2Endocrine Research
Unit, San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
This review focuses on the biologic importance of the vitamin D binding protein (DBP)
with emphasis on its regulation of total and free vitamin D metabolite levels in various
clinical conditions. Nearly all DBP is produced in the liver, where its regulation is
influenced by estrogen, glucocorticoids and inflammatory cytokines but not by vitamin
D itself. DBP is the most polymorphic protein known, and different DBP alleles can
have substantial impact on its biologic functions. The three most common alleles—Gc1f,
Gc1s, Gc2—differ in their affinity with the vitamin D metabolites and have been variably
associated with a number of clinical conditions. Although DBP has a number of biologic
functions independent of vitamin D, its major biologic function is that of regulating
circulating free and total levels of vitamin D metabolites. 25 hydroxyvitamin D (25(OH)D)
is the best studied form of vitamin D as it provides the best measure of vitamin D status.
In a normal non-pregnant individual, approximately 0.03% of 25(OH)D is free; 85% is
bound to DBP, 15% is bound to albumin. The free hormone hypothesis postulates that
only free 25(OH)D can enter cells. This hypothesis is supported by the observation
that mice lacking DBP, and therefore with essentially undetectable 25(OH)D levels,
do not show signs of vitamin D deficiency unless put on a vitamin D deficient diet.
Similar observations have recently been described in a family with a DBP mutation.
This hypothesis also applies to other protein bound lipophilic hormones including
glucocorticoids, sex steroids, and thyroid hormone. However, tissues expressing the
megalin/cubilin complex, such as the kidney, have the capability of taking up 25(OH)D still
bound to DBP, but most tissues rely on the free level. Attempts to calculate the free level
using affinity constants generated in a normal individual along with measurement of DBP
and total 25(OH)D have not accurately reflected directly measured free levels in a number
of clinical conditions. In this review, we examine the impact of different clinical conditions
as well as different DBP alleles on the relationship between total and free 25(OH)D, using
only data in which the free 25(OH)D level was directly measured. The major conclusion
is that a number of clinical conditions alter this relationship, raising the question whether
measuring just total 25(OH)D might be misleading regarding the assessment of vitamin
D status, and such assessment might be improved by measuring free 25(OH)D instead
of or in addition to total 25(OH)D.
Keywords: vitamin D binding protein, vitamin D, free 25(OH)D, free hormone hypothesis, megalin, polymorphisms,
liver cirrhosis, pregnancy
27
Bikle and Schwartz DBP and Free 25(OH)D
INTRODUCTION
Vitamin D enters the body either from its production in the
skin or absorption from the intestine. In either case, vitamin
D must be transported to tissues such as the liver where it
is metabolized to its major circulating form, 25(OH)D, by a
variety of enzymes with 25-hydroxylase activity, the major one
being CYP2R1. 25(OH)D is then transported to tissues such as
the kidney where it gets further metabolized to its biologically
active metabolite 1,25 dihydroxyvitamin D (1,25(OH)2D) by the
mitochondrial based CYP27B1. CYP24A1, found in most tissues,
is the major enzyme catabolizing 1,25(OH)2D, thus controlling
its impact on a cell specific basis. Vitamin D binding protein
(DBP) is the key transport protein which, along with albumin,
binds over 99% of the circulating vitamin D metabolites. For most
cells it is the unbound 25(OH)D that enters cells (free hormone
hypothesis), but at least in some cells such as in the kidney, and
likely in the parathyroid gland and placenta, DBP participates in
the transport of the 25(OH)D into the cell via a megalin/cubilin
complex. Although our focus will be on the transport function of
DBP and how that relates to the total and free vitamin D levels
in different physiologic and pathophysiologic conditions, DBP
has a number of functions independent of its role as a vitamin
D transport protein. These functions will be briefly reviewed as
they do contribute to the role DBP plays in health and sickness
independent of its role in vitamin D transport. DBP is a highly
polymorphic protein with at least 120 isoforms distinguished by
electrophoresis. Of these, three major isoforms have received the
most interest—Gc1f, Gc1s, and Gc2. Their structural differences
affect DBP function in ways that have an impact on a number of
clinical conditions that will be reviewed.
VITAMIN D BINDING PROTEIN
Genomic Regulation
The human DBP gene is located on chromosome 4q12-q13. It
is 35 kb in length and comprised of 13 exons encoding 474
amino acids including a 16 amino acid leader sequence, which
is cleaved before release. Numerous tissues express DBP, but the
liver is the major source (1). The expression of DBP is increased
by estrogen (2) as appreciated with the rise in DBP during
pregnancy (3,4) and with oral contraceptive administration (5).
However, the exact mechanism for this induction is not clear
as a response element for the estrogen receptor in the DBP
promoter has not been identified. Androgens, on the other hand,
do not appear to affect DBP expression (2). Dexamethasone and
certain cytokines such as IL-6 also increase DBP production,
whereas TGFβis inhibitory (6). As for estrogen, the mechanism
underlying such regulation is unclear. However, these cytokines
and glucocorticoids are likely to play a role in the increase
in DBP production following trauma (after an initial decrease
in levels due to actin clearance, see below) (7) and acute
liver failure (8), which we will discuss subsequently. Primary
hyperparathyroidism, on the other hand, is associated with
a reduction in DBP levels, likely contributing to the lower
25(OH)D levels in these patients as the free 25(OH)D is not
reduced (9). Vitamin D itself or any of its metabolites do not
regulate DBP production (10).
Structure and Polymorphisms
The mature human DBP is approximately 58 kD in size, although
differences in glycosylation of the protein for different alleles
alter the actual size. DBP is the most polymorphic gene known.
Before the appreciation of its role as a carrier of the vitamin
D metabolites these polymorphisms in DBP were used by
population geneticists to track different populations, referring
to the protein as Gc globulin. Over 120 variants have been
described based on electrophoretic properties (11) as noted above
with 1,242 polymorphisms currently listed in the NCBI database
(12). Of these variants, the Gc1f and Gc1s (rs7041 locus) and
Gc2 (rs4588 locus) are the most common (Figure 1). Gc1f and
Gc1s involve two polymorphisms, one at aa 432 (416 in the
mature DBP) and one at 436 (420 in the mature DBP). The
1f allele encodes the sequence of aa between 432 and 436 as
DATPT, the 1s allele encodes the sequence EATPT. This subtle
difference in charge makes Gcf run faster (fast) than the Gcs
(slow) during electrophoresis. The Gc2 allele encodes DATPK
which runs slower still. Glycosylation further distinguishes the
Gc1 variants from the Gc2 variant. The threonine (T) in Gc1
binds N-acetylgalactosamine to which galactose and sialic acid
bind in tandem. The lysine (K) in comparable position in Gc2
is not glycosylated (13,14). This affects the conversion of DBP
to DBP-MAF (macrophage activating factor), which involves a
partial deglycosylation removing the galactose and sialic acid by
the sequential action of sialidase and β-galactosidase by T and
B cells (15). The significance of this for the biologic function is
described below.
DBP is comprised of 3 structurally similar domains. The first
domain is the binding site for the vitamin D metabolites (aa 35–
49). Fatty acid binding utilizes a single high affinity site for both
palmitic acid and arachidonic acid, but only arachidonic acid
competes with 25(OH)D for binding (16,17). The actin binding
site is located at aa 373–403, spanning parts of domains 2 and
3, but part of domain 1 is also involved (18,19). The C5a/C5a
des Arg binding site is located at aa 130–149 (20). DBP serves
as a cochemotactic factor for C51/C5a des Arg in its regulation
of neutrophil functions (21). Membrane binding sites have been
identified in aa 150–172 and 379–402 (22).
Biologic Function
Binding to and Transport of Vitamin D Metabolites
DBP was discovered by Hirschfeld in 1959 (23), and originally
called group specific component (Gc-globulin), but it was not
until 1975 that its function as a vitamin D transport protein
was appreciated (24). In normal individuals, 85% of circulating
vitamin D metabolites are bound to DBP. Albumin binds
15% of these metabolites and does so with much lower
affinity. Approximately 0.4% of total 1,25(OH)2D3and 0.03%
of total 25OHD3are free in serum from normal non-pregnant
individuals. The affinity of DBP for the vitamin D2metabolites
is somewhat less than that for the vitamin D3metabolites (25).
The designation of “bioavailable” vitamin D metabolite is the sum
of the free vitamin D metabolite and that bound to albumin,
Frontiers in Endocrinology | www.frontiersin.org 2May 2019 | Volume 10 | Article 317
28
Bikle and Schwartz DBP and Free 25(OH)D
FIGURE 1 | The major DBP alleles. The amino acid differences between the three major DBP alleles are depicted. These differences affect not only their
electrophoretic properties but also their glycosylation pattern. In particular Gc2 is not glycosylated, which prevents it from forming the DBP-macrophage activating
factor (DBP-MAF). Other biologic differences are discussed in the text.
thus measuring around 15% in normal individuals [review in
(26)]. However, the degree to which the albumin fraction is
truly bioavailable is not clear (27). The free hormone hypothesis
postulates that only the non-bound fraction (the free fraction)
of hormones that otherwise circulate in blood bound to their
carrier proteins is able to enter cells and exert their biologic
effects. However, at least for some tissues, a transport system
has been identified that takes up the 25(OH)D (and presumably
other vitamin D metabolites) attached to DBP. That system
involves megalin/cubilin.
The role of megalin for vitamin D metabolism was discovered
by Nykjaer et al. (28), who found extensive loss of DBP in
the megalin knockout mouse and 25(OH)D in its urine. These
mice have very poor survival rates. More recently, a kidney
specific knockout of megalin was developed with a good survival
rate, enabling longer term studies that demonstrated reduced
circulating levels of the vitamin D metabolites, hypocalcemia,
and osteomalacia (29). Cubilin, together with megalin, forms
part of the complex facilitating this transport mechanism [review
in (30)]. Other tissues express the megalin/cubilin complex
including the parathyroid gland and placenta, but its role
outside the kidney has received little interest (30). Moreover,
activated monocytes may be able to accumulate DBP by a
megalin independent process, although this too needs further
study (31,32).
The physiologic role of DBP is well-illustrated in the DBP
knockout mouse. In these mice the vitamin D metabolites are
presumably all free and/or bioavailable as albumin levels are
normal. Unlike the megalin knockout mice, mice lacking DBP
do not show evidence of vitamin D deficiency unless placed
on a vitamin D deficient diet despite having very low levels of
serum 25(OH)D and 1,25(OH)2D and increased loss of these
metabolites in the urine (33). Tissue levels of 1,25(OH)2D were
normal in the DBP knockout mice, and markers of vitamin
D function such as expression of intestinal TRPV6, calbindin
9k, PMCA1b, and renal TRPV5 were maintained. Moreover,
injection of 1,25(OH)2D into these DBP knockouts showed a
more rapid increase in the expression of Cyp24A1, TRPV5,
and TRPV6 than in DBP intact controls (34). However, on
a vitamin D deficient diet they quickly developed vitamin D
deficiency. More recently, a family has been described to have
a mutation in the DBP gene deleting it from the homozygous
patient and decreasing its concentration to 50% of normal
in a heterozygous sibling (35). The homozygous patient had
nearly undetectable levels of total 25(OH)D, although the free
concentration measured directly was comparable to that of
the normal sibling, as was that of the heterozygote sibling.
Parathyroid hormone, calcium, and phosphate were all normal.
Thus, DBP does not appear necessary for getting the vitamin D
metabolites into cells, supporting the free hormone hypothesis,
but DBP clearly serves as a critical reservoir for the vitamin
D metabolites, reducing the risk of vitamin D deficiency when
intake or epidermal production is limited.
The DBP alleles have been reported to differ in their affinity
to 25(OH)D. Gc1f was initially reported as having the highest
affinity and Gc2 the lowest among the common alleles (36),
but results from other laboratories have not confirmed these
differences, and the results from later studies themselves are
inconsistent (37,38). In one such study evaluating the half life of
25(OH)D in serum, subjects homozygous for the Gc1f allele were
found to have the shortest half life indicating a reduced affinity
(39). On the other hand, serum containing the Gc1f variant of
DBP reduced the ability of 25(OH)D and 1,25(OH)2D to induce
cathelicidin in monocytes more than that of serum with the Gc2
allele, suggesting the opposite order of affinity (31). Schwartz
et al. (40) recently reported that DBP haplotype had significant
effects on total 25(OH)D, free 25(OH)D, and DBP levels. The
lowest total and free levels of 25(OH)D were seen with the Gc
2/2 haplotype which also tends to have the lowest DBP levels.
Other studies have also found lower total 25(OH)D levels in
subjects with the Gc2 allele (4145). The reason the Gc2 allele
is associated with lower DBP levels is unknown. DBP haplotype
also affected percent free 25(OH)D. The lowest free percentage
was seen with the 1s/1s haplotype and the highest one with the
Frontiers in Endocrinology | www.frontiersin.org 3May 2019 | Volume 10 | Article 317
29
Bikle and Schwartz DBP and Free 25(OH)D
1f/1f haplotype, suggesting that in this survey the Gc1s allele
had a higher affinity for 25(OH)D than the Gc1f allele, with
the Gc2 allele in between. Furthermore, the different Gc alleles
affect the response to vitamin D supplementation. Individuals
with the Gc2 variant have been shown to respond to vitamin
D supplementation with a more robust increase in 25(OH)D
(46). Moreover, within the Gc2 polymorphic region (rs4588),
individuals in an Iranian population with an AA genotype within
this polymorphic region showed a greater increase in 25(OH)D
levels following vitamin D supplementation than those with
the GG genotype did (47). Similar results were found with a
different polymorphism at rs2282679 in Caucasian women (48).
Rs2282679, an intronic polymorphism in the DBP gene that
does not alter DBP structure, was previously shown in GWAS
studies to be associated with lower 25(OH)D and DBP levels in
several different populations (4951). The clinical significance
of these allelic differences is unclear. Differences in these alleles
were not found to contribute to a difference in fracture rate
in a large study including African Americans and Caucasians
(52) or other calcemic and cardiometabolic diseases in the
Canadian Multicentre Osteoporosis Study (50). However, as
reviewed by Malik et al. (13) and Speeckaert et al. (53), a large
number of chronic diseases including type 1 and 2 diabetes
(5456), osteoporosis (5759), chronic obstructive lung disease
(60), endometriosis (61), inflammatory bowel disease (62),
some cancers (6366) [although see (6668)], and tuberculosis
(69) have been associated with DBP variants. Other SNPs at
rs4588 have been associated with susceptibility to the metabolic
syndrome (70). At the Gc1 locus (rs7041) the G allele is associated
with increased susceptibility to hepatitis C viral infection (71).
Karras et al. (72) has summarized a number of studies showing
the impact of DBP and DBP polymorphisms on various outcomes
of pregnancy. These studies demonstrate the recent interest in
the impact of polymorphisms on DBP function, but it remains
to be seen whether these initial results will be generalized across
different populations.
Actin Scavenging
A major function of DBP that has received considerably less
interest than that of vitamin D metabolite binding is its role
in actin scavenging. Following trauma (7), sepsis (7375), liver
trauma (8,76,77), acute lung injury (78), preeclampsia (79),
surgery (80,81), and burn injuries (82), large amounts of
actin are released from the damaged cells forming polymerized
filamentous F-actin that, in combination with coagulation factor
Va, can lead to disseminated intravascular coagulation and
multiorgan failure unless cleared (83). The actin scavenging
system consists of gelsolin and DBP. Gelsolin depolymerizes F-
actin to G (globular) actin. DBP, with its high affinity for G-actin
(Kd =10 nM), prevents the repolymerization and clears it from
the blood (84,85). No clear difference among the major DBP
variants has been observed regarding binding to G-actin (53).
The DBP-actin complexes are rapidly cleared (half life in blood
approximately 30 min) (81), primarily by the liver, lungs and
spleen. These tissues have receptors for the DBP-actin complexes
(86). The acute conditions result in a fall in DBP levels, potentially
decreasing the bioavailability of the vitamin D metabolites (8,87,
88), with a rise in the DBP-actin complexes (7,73,77,78). The
ability of the organism to respond to the insult by increasing DBP
production is correlated to survival (7,8,89), and has led to the
consideration of the use of DBP therapeutically (90,91).
Neutrophil Recruitment and Migration With
Complement 5a (C5a) Binding
Neutrophil activation during inflammation increases their
binding sites for DBP (92), and DBP binding to these sites
facilitates C5a induced chemotaxis (21) as well as other
chemoattractants such as CXCL1 during inflammation (93).The
interaction with C5a involves residues 130–149 of DBP, a region
which is common to all major DBP alleles (20), and no difference
in these alleles has been found with respect to their promotion of
C5a mediated chemotaxis (21). Binding of 1,25(OH)2D but not
25(OH)D blocks the promotion by DBP of C5a activity (94).
Fatty Acid Binding
DBP binds fatty acids but with lower affinity (Ka =105-106M1)
than albumin and via a single binding site (16,95). Most of the
fatty acids binding to DBP are mono-unsaturated or saturated,
with only 5% poly-unsaturated. However, only poly-unsaturated
fatty acids such as arachidonic acid and linoleic acid compete
with vitamin D metabolites for DBP binding (17,96). This
suggests that the different fatty acids alter the configuration
of DBP affecting the binding of the vitamin D metabolites
rather than directly competing with the vitamin D metabolites
for their binding site. The role of DBP in fatty acid transport
appears limited.
Formation of the DBP-Macrophage Activating Factor
(DBP-MAF) and its Functions
As described above, DBP-MAF is formed from certain alleles
(Gc1s and 1f) of DBP following deglycoslyation during
inflammatory processes (97). These deglycosylation steps are
required for the role of DBP in macrophage activation (15), but
further removal of the N-acetyl-galactosamine (NaGal) reduces
this activity (98). DBP-MAF is able to activate osteoclasts (99)
independent of its 25(OH)D binding function, and it has been
shown to stimulate bone resorption in the osteopetrosis (OP)
and the incisor absent (IA) rat (100). DBP-MAF has also shown
efficacy in a number of tumor models (101103). Removal of
NaGal by α-NaGalase blocks DBP-MAF formation contributing
to the loss of immunosuppression in cancer patients (104). α-
NaGalase is produced in the liver, and appears to be directly
related to tumor burden (105). Preparations of DBP-MAF may
have therapeutic potential (14).
FREE HORMONE HYPOTHESIS
As previously noted, the free hormone hypothesis postulates that
only the non-bound fraction (the free fraction) of hormones
that otherwise circulates in blood bound to their carrier proteins
is able to enter cells and exert their biologic effects (Figure 2).
Examples include the vitamin D metabolites, which we are
discussing in this review, sex steroids, cortisol, and thyroid
hormone. These are lipophilic hormones assumed to cross
Frontiers in Endocrinology | www.frontiersin.org 4May 2019 | Volume 10 | Article 317
30
Bikle and Schwartz DBP and Free 25(OH)D
FIGURE 2 | The Free Vitamin D hypothesis. As noted in the text, vitamin D (OH) metabolites are bound to D Binding Protein (DBP) and to a lesser extent albumin in
the circulation. These cross the cell membrane as the free (unbound) metabolite in most tissue. However, In the kidney, parathyroid gland, and placenta, the
megalin/cubilin complex can transport bound D (OH) metabolites into cells.
the plasma membrane by diffusion and not by an active
transport mechanism. One of the earliest clinical examples
leading to the formulation of the free hormone hypothesis
came from observations by Recant and Riggs (106) that patients
with protein losing nephropathy developed quite low levels of
thyroid hormone (PBI) along with increased urinary losses but
without evidence of hypothyroidism. Subsequent studies have
established the free hormone hypothesis for the thyroid and
steroid hormones (107,108), and measurements of the free
concentrations of thyroid hormone, estrogen, and testosterone
are standard practice. As will be discussed subsequently, this is
likely to become the case for free 25(OH)D. As noted earlier,
mice lacking DBP lost substantial amounts of the vitamin
D metabolites in the urine with marked reductions in their
circulating levels of 25(OH) D, but they did not develop evidence
of rickets until put on a low vitamin D diet. Such results indicate
the importance of the free fraction of 25(OH)D for biologic
functions and the role of DBP as a circulating reservoir (33).
To address the clinical relevance of the free hormone
hypothesis for vitamin D metabolites, a method to measure
the free concentration needed to be developed. This was
originally performed by centrifugal ultrafiltration to directly
determine the free levels of 25(OH)D and 1,25(OH)2D (109,
110) in various clinical situations. However, this method is
labor intensive and has recently been replaced at least for
free 25(OH)D by a two-step ELISA that directly measures
free 25(OH)D (Future Diagnostics Solutions B.V., Wijchen,
Netherlands) using monoclonal antibodies from DIAsource
Immunoassays (Louvain-la-Neuve, Belgium). The antibody in
the current assay does not recognize 25(OH)D2as well as
25(OH)D3(77% of the 25(OH)D3value), so underestimates
the free 25(OH)D2. However, under most situations where
the predominant vitamin D metabolite is 25(OH)D3, the data
compare quite well to those obtained from similar populations
using the centrifugal ultrafiltration assay (111,112). The initial
studies with the centrifugal ultrafiltration method established
affinity constants for DBP and albumin binding to 25(OH)D and
1,25(OH)2D in a healthy young adult (DD Bikle) and may not
be generalizable to a broad range of individuals from different
ethnic backgrounds or in different clinical conditions. However,
prior to the development of a high throughput ELISA assay to
measure the free concentration directly, these affinity constants
proved useful in calculating the free concentrations (113,114)
from measurements of DBP, albumin and the total vitamin D
metabolite of interest according to the formula:
free vitamin D metabolite =total vitamin D metabolite
1+Kaalbalbumin+(KaDBPDBP)
As noted previously, the affinity of 25(OH)D for albumin is
much less that than for DBP, leading some to consider albumin-
bound 25(OHD) to be essentially “free” or “available” and define
“bioavailable 25(OH)D” as free 25(OH)D plus albumin-bound
25(OH)D. Given that the albumin bound 25(OH)D (15%) is
considerably higher than the free level (0.03%), this would imply
that approximately 500 times as much 25(OH)D is available to
cells than if only the free fractions were available. There is little
evidence to support albumin bound 25(OH)D as being readily
available to cells.
Frontiers in Endocrinology | www.frontiersin.org 5May 2019 | Volume 10 | Article 317
31
Bikle and Schwartz DBP and Free 25(OH)D
In sera from normal healthy younger individuals, the
calculated values of free 25(OH)D and 1,25(OH)2D using
DBP measured with polyclonal antibodies correlate reasonably
well with the directly measured free levels using centrifugal
ultrafiltration for both metabolites or the ELISA assay for
25(OH)D. However, when applied to clinical populations with
altered DBP levels either during physiologic (e.g., pregnancy)
or pathologic (eg. liver disease) conditions, the calculated values
no longer are consistent with those measured directly by either
centrifugal ultrafiltration or the newly developed ELISA (115).
Part of this is due to the disparity between assays for both
the vitamin D metabolite (e.g., 25(OH)D) and DBP, each of
which have generally relied on immunoassays. However, mass
spectroscopy is becoming the gold standard for measurement
of the vitamin D metabolites (116,117) and is being developed
for the measurement of DBP and its various isoforms as well
(42,118). The adoption of mass spectroscopy should reduce the
variation in these measurements from different laboratories. But
a major problem in attempting to calculate the free fraction of
vitamin D metabolites is the assumption that all DBP alleles
have the same affinity for the vitamin D metabolites, and that
this is invariant under varying clinical conditions. As noted
previously, the rank order of affinity of the different alleles for the
vitamin D metabolites remains controversial, but differences have
been found. Regardless, these potential differences in measured
affinity do not begin to explain the large differences between the
calculated and directly measured free metabolite levels in various
disease states (40). Although there are statistically significant
correlations between calculated and directly measured free
25(OH)D, the relationship accounts for only 13% of the variation.
Calculated free 25(OH)D concentrations are consistently higher
than directly measured concentrations in a variety of studies,
such as those performed during the third trimester of pregnancy
and in patients with liver disease or cystic fibrosis (115,119
122). These studies suggest changes in the affinity of 25(OH)D
to DBP independent of allelic variations in at least some of these
clinical conditions.
CLINICAL STUDIES
Healthy Populations
Determinations of free 25(OH)D concentrations in healthy
populations show highly significant correlations with total
25(OH)D concentrations whether measured directly or
indirectly. Assays to directly measure free 25(OH)D are not
currently available for use in clinical care but have been used
in research investigations. As noted above, calculated 25(OH)D
values are usually higher than when measured directly, which
is based on multiple unsubstantiated assumptions such that
results obtained with the two methods can differ markedly in
different clinical conditions. For these reasons only results from
studies with directly measured free 25(OH)D will be discussed.
When measured with the direct immunoassay, free 25(OH)D
levels have been reported to be between 0.02 and 0.09% of
total 25(OH)D concentrations and generally range from 0.5
to 8.1 pg/mL in 95% of healthy adults (Figure 3). However,
clinical conditions that alter either DBP, the affinity of DBP for
FIGURE 3 | Distribution of free 25(OH)D in Adults and Selected Patient
Groups. Distribution of directly-measured free 25(OH)D in normal adults (in
green), pregnant women (pink), cirrhotics (orange), and nursing home
residents (gray). Distributions are shifted leftward toward lower free 25(OH)D
concentrations in pregnant women in the 2nd and 3rd trimesters concordant
with increased DBP while decreased synthetic function and DBP in cirrhotics
shifts free 25(OH) concentrations to the right toward higher levels. The
mechanism for higher free 25(OH) concentrations in Nursing home residents is
likely related to D supplementation, somewhat lower, albumin, and the
pro-inflammatory state of frailty. Figure generated form data in
Schwartz et al. (40).
25(OH)D metabolites or albumin, or disposition of vitamin
D, may alter free 25(OH)D concentrations or relationships
between free and total 25(OH)D concentrations. In this regard,
a number of medications, hormones, and smoking have been
shown to affect DBP levels (123). Thus, as shown in Figure 3, the
free concentration of 25(OH)D varies among different clinical
conditions. DBP haplotypes have also been hypothesized to
alter the affinity between total 25(OH)D and free 25(OH)D,
although, as shown in Figure 4, the variation in percent
free 25(OH)D levels is less affected by DBP haplotype than
clinical condition.
Free 25(OH) D in Conditions That Alter DBP
Pregnancy
As pregnancy progresses there are time dependent changes
in DBP with almost two-fold increases between the second
and third trimesters. Despite these marked DBP changes,
mean free 25(OH)D may be the same as or only slightly lower
than in non-pregnant women but with less variability than
in other groups (40,124). The slope of the free 25(OH)D
vs. total 25(OH) D relationship, however, is significantly
less steep than in healthy individuals. The same conclusion
was drawn from earlier studies with measurements of free
1,25(OH)2D (109). These results suggest that the affinity
of DBP for vitamin D metabolites is decreased during
pregnancy, perhaps compensating for increased DBP
Frontiers in Endocrinology | www.frontiersin.org 6May 2019 | Volume 10 | Article 317
32
Bikle and Schwartz DBP and Free 25(OH)D
FIGURE 4 | Percent free 25(OH)D in adults by clinical condition or DBP Haplotype. Percent free 25(OH)D concentrations for selected clinical groups on the left panel
(community outpatients, NH=nursing home patients, cirrhotics, pregnant women, prediabetics, and normal individuals) and by DBP haplotype on the right . Boxplots
show 10th, 25th, median, 75th, and 90th percentile values. Individual points represent values above the 90th and below the 10th percentiles. Both clinical subgroup
and DBP genotype significantly effect percentage free 25(OH)D. Between group comparisons for clinical conditions were significant for all but healthy persons
compared with pregnant women or outpatients, or for pregnant women compared with outpatients. For DBP haplotypes, smaller but significant differences were
detected between the 1s/1s haplotype and the 1s/1f,1f/2, 1f/1f, and 1s/2 haplotypes and between the 1s/2 and 1f/2 and 1f/1f haplotypes and between the 1s/1f and
1f/1f haplotypes. Data are reproduced with permission from Schwartz et al. (40).
concentrations and the needs of both the mother and fetus
for calcium.
Liver Disease
Liver diseases that are associated with impaired protein synthetic
function such as cirrhosis and acute liver failure result in
reductions in DBP and albumin. In addition, the relationship
between free 25(OH)D and total 25(OH)D is significantly steeper
in patients with cirrhosis than in healthy people indicating altered
affinity of DBP for 25(OH)D (40) (Figure 4). The net result
is that directly measured free 25(OH)D is higher and shows
greater variability in patients with cirrhosis compared to healthy
individuals and stable outpatients with other chronic conditions
(40,110,115) despite lower total 25(OH) D concentrations.
Results regarding the effects of cirrhosis or acute liver failure
on the relationship of total to free 25(OH)D are consistent,
creating a strong argument for assessment of free 25(OH)D to
assess vitamin D status in the presence of liver pathology as total
25(OH)D measurements may be misleading.
Renal Disease
Nephrotic syndrome, acute renal failure, acute tubular necrosis,
or chronic kidney disease associated with renal tubular necrosis
may have decreased transport capacity for DBP from the
glomerular filtrate into the renal tubules. Heavy proteinuria
can lead to loss of DBP as well as 25(OH)D in the urine as
the maximal transport capacity of the megalin/cubulin system
is saturated. Reports in the literature have not included direct
measurement of free 25(OH)D in these conditions, but a small
study of nephrotics showed lower total and free 1,25(OH)2D
compared to people with normal renal function (125).
Clinical Conditions Not Associated With
Altered DBP Levels
Obesity
High BMIs are associated with reductions in total and free
25(OH)D but not DBP or elimination of half-life measurements
of 25(OH)D (126). The underlying mechanism for these changes
is unknown but may be related to the pro-inflammatory state
and circulating cytokines present in obesity, although increased
volume of distribution (into fat) has also been invoked.
DBP Haplotypes
Investigations using direct measurements of free 25(OH)D have
detected statistically significant but not marked differences in free
25(OH)D concentrations between healthy individuals with the
six common DBP haplotypes (Figure 4). This is in contrast to the
marked differences between haplotypes reported with calculated
free 25(OH)D levels (122,127). As noted previously with directly
measured free 25(OH)D, the lowest free 25(OH)D is seen with
the Gc 2/2 haplotype and the highest levels with the 1s alleles. Per
cent free was highest with the 1f/1f haplotype in our studies (40)
(see Figure 4).
Nursing Home Subjects
In a vitamin D dose titration study (128) of nursing home
residents, who are older, have more chronic co-morbidities, and
receive more medications than younger people or community-
dwelling elderly, free 25(OH)D levels rose along with increases
in total 25(OH)D. The per cent free was higher than in younger
adults. Relationships between free and total 25(OH)D were also
steeper than those of normal subjects or younger outpatients
suggesting altered affinity of 25(OH)D to DBP in this group.
Slightly lower albumin concentrations may have also had a
small contribution. Inflammation and/or elevated cytokines that
Frontiers in Endocrinology | www.frontiersin.org 7May 2019 | Volume 10 | Article 317
33
Bikle and Schwartz DBP and Free 25(OH)D
accompany very old age or multiple morbidities may have
also contributed to altered affinity of 25(OH)D to DBP in this
group (129).
Associations With Markers of Vitamin D Biologic
Function
PTH is generally found to be negatively correlated with
free 25(OH)D as well as total 25(OH)D. Reports variably
conclude that one or the other shows a slightly more
significant relationship, but neither explains more than a small
amount of the variability in the relationship. Moreover, if
the megalin/cubilin complex is operative in the parathyroid
gland as it is in the kidney, PTH levels may not be able to
distinguish between free and total 25(OH)D with respect to
biologic action. However, further insight into the impact of free
vs. total 25(OH)D on PTH levels may be gained from several
recent studies showing that with high dose D supplementation,
changes in iPTH were significantly related to changes in directly
measured free 25(OH)D but not to changes in total 25(OH)D
(128,130,131), suggesting that free 25(OH)D might be a better
marker of the biologically available fraction at higher total
25(OH)D concentrations or when 25(OH)D is changing. Data on
relationships between directly measured free 25(OH)D and bone
density or markers of bone turnover are inconsistent.
Other Conditions
There are limited data on the effect of oral contraceptives or
hormone replacement therapy with estrogen, but free 25(OH)D
levels and relationships between total and free 25(OH)D do not
appear to be significantly influenced by the use of these agents
at currently prescribed dosages and routes of administration.
Similarly, stable medical conditions such as hypertension,
prediabetes, diabetes, osteoporosis, or mild renal disease do
not appear to significantly alter relationships between free and
total 25(OH)D.
Summary of Clinical Studies
The impact of clinical conditions on free 25(OH)D is that the
absolute level, the percent free 25(OH)D and the relationship
between free and total 25(OH)D concentrations, differ in
pregnant women, 336 people with cirrhosis, and elderly people
with multiple morbidities compared to normals or community-
dwelling outpatients. These relationships are affected to a much
smaller extent by BMI in all groups. It is key that while DBP
haplotype variation is associated with differences in per cent free
25(OH)D, the DBP haplotype effects are far smaller in magnitude
than those of pregnancy, cirrhosis, or very old nursing home
residents with multiple chronic conditions. Thus, total 25(OH)D
measurements may be misleading in persons with altered total-
to-free relationships, although for other clinical conditions
the relationship between total and free 25(OH)D may be
less affected.
CONTRIBUTION TO THE FIELD
25(OH)D measurements in the blood currently provide the
standard assessment of vitamin D status. Nearly all 25(OH)D
circulates as the bound form, with the vitamin D binding protein
(DBP) accounting for approximately 85% of the binding, with
albumin accounting for most of the rest. However, it is the very
small percentage that is not protein bound (0.03% in normal
individuals) that is able to cross the membrane of most cells.
Conditions that alter levels of DBP or its binding to 25(OH)D
alter the relationship between free and total levels. If the free
concentration provides a more accurate assessment of vitamin D
status, measuring only total 25(OH)D levels may be misleading
in situations where the relationship between total and free
25(OH)D levels is altered as in liver disease and pregnancy or in
individuals with different DBP alleles. This review examines the
impact of different DBP alleles and clinical conditions that do the
relationship between free and total 25(OH)D levels, concluding
that in a number of clinical situations measuring the free level
may provide a better index of vitamin D status than total levels in
such situations.
AUTHOR CONTRIBUTIONS
All authors listed have made a substantial, direct and intellectual
contribution to the work, and approved it for publication.
FUNDING
Grant support provided by: NIH AR 055924 (DB), VA
I01BX003814 (DB).
ACKNOWLEDGMENTS
We appreciate the provision of data by our coauthors in our
publication Schwartz et al. (40), that is included in this review.
REFERENCES
1. Cooke NE, McLeod JF, Wang XK, Ray K. Vitamin D binding
protein: genomic structure, functional domains, and mRNA
expression in tissues. J Steroid Biochem Mol Biol. (1991)
40:787–93. doi: 10.1016/0960-0760(91)90304-N
2. Hagenfeldt Y, Carlstrom K, Berlin T, Stege R. Effects of orchidectomy
and different modes of high dose estrogen treatment on circulating “free”
and total 1,25-dihydroxyvitamin D in patients with prostatic cancer. J
Steroid Biochem Mol Biol. (1991) 39:155–9. doi: 10.1016/0960-0760(91)
90056-B
3. Moller UK, Streym S, Heickendorff L, Mosekilde L, Rejnmark L. Effects of
25OHD concentrations on chances of pregnancy and pregnancy outcomes:
a cohort study in healthy Danish women. Eur J Clin Nutr. (2012) 66:862–
8. doi: 10.1038/ejcn.2012.18
4. Zhang JY, Lucey AJ, Horgan R, Kenny LC, Kiely M. Impact of pregnancy
on vitamin D status: a longitudinal study. Br J Nutr. (2014) 112:1081–
7. doi: 10.1017/S0007114514001883
5. Moller UK, Streym S, Jensen LT, Mosekilde L, Schoenmakers I, Nigdikar
S, et al. Increased plasma concentrations of vitamin D metabolites and
vitamin D binding protein in women using hormonal contraceptives: a
cross-sectional study. Nutrients. (2013) 5:3470–80. doi: 10.3390/nu5093470
Frontiers in Endocrinology | www.frontiersin.org 8May 2019 | Volume 10 | Article 317
34
Bikle and Schwartz DBP and Free 25(OH)D
6. Guha C, Osawa M, Werner PA, Galbraith RM, Paddock GV. Regulation
of human Gc (vitamin D–binding) protein levels: hormonal and
cytokine control of gene expression in vitro.Hepatology. (1995)
21:1675–81. doi: 10.1016/0270-9139(95)90474-3
7. Dahl B, Schiodt FV, Rudolph S, Ott P, Kiaer T, Heslet L. Trauma
stimulates the synthesis of Gc-globulin. Intens Care Med. (2001) 27:394–
9. doi: 10.1007/s001340000837
8. Schiodt FV. Gc-globulin in liver disease. Dan Med Bull. (2008) 55:131–46.
9. Wang X, Shapses SA, Al-Hraishawi H. Free and bioavailable 25-
Hydroxyvitamin D levels in patients with primary hyperparathyroidism.
Endocr Pract. (2017) 23:66–71. doi: 10.4158/EP161434.OR
10. Bjorkhem-Bergman L, Torefalk E, Ekstrom L, Bergman P. Vitamin D
binding protein is not affected by high-dose vitamin D supplementation: a
post hoc analysis of a randomised, placebo-controlled study. BMC Res Notes.
(2018) 11:619. doi: 10.1186/s13104-018-3725-7
11. Cleve H, Constans J. The mutants of the vitamin-D-binding protein:
more than 120 variants of the GC/DBP system. Vox Sang. (1988) 54:215–
25. doi: 10.1111/j.1423-0410.1988.tb03908.x
12. Chun RF. New perspectives on the vitamin D binding protein. Cell Biochem
Funct. (2012) 30:445–56. doi: 10.1002/cbf.2835
13. Malik S, Fu L, Juras DJ, Karmali M, Wong BY, Gozdzik A, et al. Common
variants of the vitamin D binding protein gene and adverse health outcomes.
Crit Rev Clin Lab Sci. (2013) 50:1–22. doi: 10.3109/10408363.2012.750262
14. Nagasawa H, Uto Y, Sasaki H, Okamura N, Murakami A, Kubo S, et al. Gc
protein (vitamin D-binding protein): Gc genotyping and GcMAF precursor
activity. Anticancer Res. (2005) 25:3689–95.
15. Uto Y, Yamamoto S, Mukai H, Ishiyama N, Takeuchi R, Nakagawa Y,
et al. Beta-Galactosidase treatment is a common first-stage modification
of the three major subtypes of Gc protein to GcMAF. Anticancer Res.
(2012) 32:2359–64.
16. Calvo M, Ena JM. Relations between vitamin D and fatty acid binding
properties of vitamin D-binding protein. Biochem Biophys Res Commun.
(1989) 163:14–7. doi: 10.1016/0006-291X(89)92091-3
17. Bouillon R, Xiang DZ, Convents R, Van Baelen H. Polyunsaturated fatty
acids decrease the apparent affinity of vitamin D metabolites for human
vitamin D-binding protein. J Steroid Biochem Mol Biol. (1992) 42:855–
61. doi: 10.1016/0960-0760(92)90094-Y
18. Haddad JG, Hu YZ, Kowalski MA, Laramore C, Ray K, Robzyk P,
et al. Identification of the sterol- and actin-binding domains of plasma
vitamin D binding protein (Gc-globulin). Biochemistry. (1992) 31:7174–
81. doi: 10.1021/bi00146a021
19. Head JF, Swamy N, Ray R. Crystal structure of the complex between actin and
human vitamin D-binding protein at 2.5 A resolution. Biochemistry. (2002)
41:9015–20. doi: 10.1021/bi026054y
20. Zhang J, Kew RR. Identification of a region in the vitamin D-binding protein
that mediates its C5a chemotactic cofactor function. J Biol Chem. (2004)
279:53282–7. doi: 10.1074/jbc.M411462200
21. Binder R, Kress A, Kan G, Herrmann K, Kirschfink M. Neutrophil priming
by cytokines and vitamin D binding protein (Gc-globulin): impact on C5a-
mediated chemotaxis, degranulation and respiratory burst. Mol Immunol.
(1999) 36:885–92. doi: 10.1016/S0161-5890(99)00110-8
22. Wilson RT, Bortner JD Jr, Roff A, Das A, Battaglioli EJ, Richie
JP Jr, et al. Genetic and environmental influences on plasma
vitamin D binding protein concentrations. Transl Res. (2015)
165:667–76. doi: 10.1016/j.trsl.2014.08.003
23. Hirschfeld J. Immune-electrophoretic demonstration of qualitative
differences in human sera and their relation to the haptoglobins. Acta Pathol
Microbiol Scand. (1959) 47:160–8. doi: 10.1111/j.1699-0463.1959.tb04844.x
24. Daiger SP, Schanfield MS, Cavalli-Sforza LL. Group-specific component (Gc)
proteins bind vitamin D and 25-hydroxyvitamin D. Proc Natl Acad Sci USA.
(1975) 72:2076–80. doi: 10.1073/pnas.72.6.2076
25. Armas LA, Hollis BW, Heaney RP. Vitamin D2 is much less effective
than vitamin D3 in humans. J Clin Endocrinol Metab. (2004) 89:5387–
91. doi: 10.1210/jc.2004-0360
26. Bikle DD, Malmstroem S, Schwartz J. Current controversies: are free
vitamin metabolite levels a more accurate assessment of vitamin D status
than total levels? Endocrinol Metab Clin North Am. (2017) 46:901–
18. doi: 10.1016/j.ecl.2017.07.013
27. Bikle DD, Gee E. Free, and not total, 1,25-dihydroxyvitamin D regulates
25-hydroxyvitamin D metabolism by keratinocytes. Endocrinology. (1989)
124:649–54. doi: 10.1210/endo-124-2-649
28. Nykjaer A, Dragun D, Walther D, Vorum H, Jacobsen C, Herz
J, et al. An endocytic pathway essential for renal uptake and
activation of the steroid 25-(OH) vitamin D3. Cell. (1999)
96:507–15. doi: 10.1016/S0092-8674(00)80655-8
29. Leheste JR, Melsen F, Wellner M, Jansen P, Schlichting U, Renner-Muller
I, et al. Hypocalcemia and osteopathy in mice with kidney-specific megalin
gene defect. FASEB J. (2003) 17:247–9. doi: 10.1096/fj.02-0578fje
30. Christensen EI, Birn H. Megalin and cubilin: multifunctional endocytic
receptors. Nat Rev Mol Cell Biol. (2002) 3:256–66. doi: 10.1038/nrm778
31. Chun RF, Lauridsen AL, Suon L, Zella LA, Pike JW, Modlin RL, et al.
Vitamin D-binding protein directs monocyte responses to 25-hydroxy-
and 1,25-dihydroxyvitamin D. J Clin Endocrinol Metab. (2010) 95:3368–
76. doi: 10.1210/jc.2010-0195
32. Esteban C, Geuskens M, Ena JM, Mishal Z, Macho A, Torres JM, et al.
Receptor-mediated uptake and processing of vitamin D-binding protein in
human B-lymphoid cells. J Biol Chem. (1992) 267:10177–83.
33. Safadi FF, Thornton P, Magiera H, Hollis BW, Gentile M, Haddad JG, et al.
Osteopathy and resistance to vitamin D toxicity in mice null for vitamin D
binding protein. J Clin Invest. (1999) 103:239–51. doi: 10.1172/JCI5244
34. 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) 149:3656–67. doi: 10.1210/en.2008-0042
35. Henderson CM, Fink SL, Bassyouni H, Argiropoulos B, Brown L, Laha TJ,
et al. Vitamin D-binding protein deficiency and homozygous deletion of the
GC gene. N Engl J Med. (2019) 380:1150–7. doi: 10.1056/NEJMoa1807841
36. Arnaud J, Constans J. Affinity differences for vitamin D metabolites
associated with the genetic isoforms of the human serum carrier protein
(DBP). Hum Genet. (1993) 92:183–8. doi: 10.1007/BF00219689
37. Bouillon R, van Baelen H, de Moor P. Comparative study of the affinity of
the serum vitamin D-binding protein. J Steroid Biochem. (1980) 13:1029–
34. doi: 10.1016/0022-4731(80)90133-8
38. Boutin B, Galbraith RM, Arnaud P. Comparative affinity of the major
genetic variants of human group-specific component (vitamin D-binding
protein) for 25-(OH) vitamin D. J Steroid Biochem. (1989) 32:59–
63. doi: 10.1016/0022-4731(89)90014-9
39. Jones KS, Assar S, Harnpanich D,Bouillon R , Lambrechts D,Prentice A, 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) 99:3373–
81. doi: 10.1210/jc.2014-1714
40. Schwartz JB, Gallagher JC, Jorde R, Berg V, Walsh J, Eastell 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) 103:3278–88. doi: 10.1210/jc.2018-00295
41. Lauridsen AL, Vestergaard P, Nexo E. Mean serum concentration of vitamin
D-binding protein (Gc globulin) is related to the Gc phenotype in women.
Clin Chem. (2001) 47:753–6.
42. Hoofnagle AN, Eckfeldt JH, Lutsey PL. Vitamin D-binding protein
concentrations quantified by mass spectrometry. N Engl J Med. (2015)
373:1480–2. doi: 10.1056/NEJMc1502602
43. Carpenter TO, Zhang JH, Parra E, Ellis BK, Simpson C, Lee WM, et al.
Vitamin D binding protein is a key determinant of 25-hydroxyvitamin
D levels in infants and toddlers. J Bone Mineral Res. (2013) 28:213–
21. doi: 10.1002/jbmr.1735
44. Santos BR, Mascarenhas LP, Boguszewski MC, Spritzer PM. Variations
in the vitamin D-binding protein (DBP) gene are related to lower 25-
hydroxyvitamin D levels in healthy girls: a cross-sectional study. Hormone
Res Paediatr. (2013) 79:162–8. doi: 10.1159/000348847
45. Shao B, Jiang S, Muyiduli X, Wang S, Mo M, Li M, et al. Vitamin D pathway
gene polymorphisms influenced vitamin D level among pregnant women.
Clin Nutr. (2018) 37:2230–7. doi: 10.1016/j.clnu.2017.10.024
46. Fu L, Yun F, Oczak M, Wong BY, Vieth R, Cole DE. Common genetic variants
of the vitamin D binding protein (DBP) predict differences in response of
serum 25-hydroxyvitamin D [25(OH)D] to vitamin D supplementation. Clin
Biochem. (2009) 42:1174–7. doi: 10.1016/j.clinbiochem.2009.03.008
Frontiers in Endocrinology | www.frontiersin.org 9May 2019 | Volume 10 | Article 317
35
Bikle and Schwartz DBP and Free 25(OH)D
47. Mehramiz M, Khayyatzadeh SS, Esmaily H, Ghasemi F, Sadeghi-Ardekani
K, Tayefi M, et al. Associations of vitamin D binding protein variants with
the vitamin D-induced increase in serum 25-hydroxyvitamin D. Clin Nutr
ESPEN. (2019) 29:59–64. doi: 10.1016/j.clnesp.2018.12.005
48. Moon RJ, Harvey NC, Cooper C, D’Angelo S, Curtis EM, Crozier SR, et al.
Response to antenatal cholecalciferol supplementation is associated with
common vitamin D-related genetic variants. J Clin Endocrinol Metab. (2017)
102:2941–9. doi: 10.1210/jc.2017-00682
49. Wang TJ, Zhang F, Richards JB, Kestenbaum B, van Meurs JB,
Berry D, et al. Common genetic determinants of vitamin D
insufficiency: a genome-wide association study. Lancet. (2010)
376:180–8. doi: 10.1016/S0140-6736(10)60588-0
50. Leong A, Rehman W, Dastani Z, Greenwood C, Timpson N, Langsetmo L,
et al. The causal effect of vitamin D binding protein (DBP) levels on calcemic
and cardiometabolic diseases: a Mendelian randomization study. PLoS Med.
(2014) 11:e1001751. doi: 10.1371/journal.pmed.1001751
51. Cheung CL, Lau KS, Sham PC, Tan KC, Kung AW. Genetic variant in
vitamin D binding protein is associated with serum 25-hydroxyvitamin D
and vitamin D insufficiency in southern Chinese. J Hum Genet. (2013)
58:749–51. doi: 10.1038/jhg.2013.84
52. Takiar R, Lutsey PL, Zhao D, Guallar E, Schneider AL, Grams ME,
et al. The associations of 25-hydroxyvitamin D levels, vitamin D binding
protein gene polymorphisms, and race with risk of incident fracture-related
hospitalization: twenty-year follow-up in a bi-ethnic cohort (the ARIC
Study). Bone. (2015) 78:94–101. doi: 10.1016/j.bone.2015.04.029
53. Speeckaert M, Huang G, Delanghe JR, Taes YE. Biological and clinical aspects
of the vitamin D binding protein (Gc-globulin) and its polymorphism. Clin
Chim Acta. (2006) 372:33–42. doi: 10.1016/j.cca.2006.03.011
54. Hirai M, Suzuki S, Hinokio Y, Chiba M, Kasuga S, Hirai A, et al.
Group specific component protein genotype is associated with
NIDDM in Japan. Diabetologia. (1998) 41:742–3. doi: 10.1007/s0012500
50980
55. Baier LJ, Dobberfuhl AM, Pratley RE, Hanson RL, Bogardus C. Variations in
the vitamin D-binding protein (Gc locus) are associated with oral glucose
tolerance in nondiabetic Pima Indians. J Clin Endocrinol Metab. (1998)
83:2993–6. doi: 10.1210/jcem.83.8.5043
56. Ye WZ, Dubois-Laforgue D, Bellanne-Chantelot C, Timsit J, Velho G.
Variations in the vitamin D-binding protein (Gc locus) and risk of type
2 diabetes mellitus in French Caucasians. Metab Clin Exp. (2001) 50:366–
9. doi: 10.1053/meta.2001.20172
57. Lauridsen AL, Vestergaard P, Hermann AP, Moller HJ, Mosekilde L, Nexo E.
Female premenopausal fracture risk is associated with gc phenotype. J Bone
Mineral Res. (2004) 19:875–81. doi: 10.1359/JBMR.040133
58. Papiha SS, Allcroft LC, Kanan RM, Francis RM, Datta HK. Vitamin D
binding protein gene in male osteoporosis: association of plasma DBP and
bone mineral density with (TAAA)(n)-Alu polymorphism in DBP. Calcified
Tissue Int. (1999) 65:262–6. doi: 10.1007/s002239900695
59. Ezura Y, Nakajima T, Kajita M, Ishida R, Inoue S, Yoshida H, et al.
Association of molecular variants, haplotypes, and linkage disequilibrium
within the human vitamin D-binding protein (DBP) gene with
postmenopausal bone mineral density. J Bone Mineral Res. (2003)
18:1642–9. doi: 10.1359/jbmr.2003.18.9.1642
60. Chishimba L, Thickett DR, Stockley RA, Wood AM. The vitamin D axis
in the lung: a key role for vitamin D-binding protein. Thorax. (2010)
65:456–62. doi: 10.1136/thx.2009.128793
61. Faserl K, Golderer G, Kremser L, Lindner H, Sarg B, Wildt L, et al.
Polymorphism in vitamin D-binding protein as a genetic risk factor in the
pathogenesis of endometriosis. J Clin Endocrinol Metab. (2011) 96:E233–
241. doi: 10.1210/jc.2010-1532
62. Eloranta JJ, Wenger C, Mwinyi J, Hiller C, Gubler C, Vavricka SR, et al.
Association of a common vitamin D-binding protein polymorphism with
inflammatory bowel disease. Pharmacogenet Genomics. (2011) 21:559–
64. doi: 10.1097/FPC.0b013e328348f70c
63. Abbas S, Linseisen J, Slanger T, Kropp S, Mutschelknauss EJ, Flesch-Janys
D, et al. The Gc2 allele of the vitamin D binding protein is associated
with a decreased postmenopausal breast cancer risk, independent of the
vitamin D status. Cancer Epidemiol Biomarkers Prev. (2008) 17:1339–
43. doi: 10.1158/1055-9965.EPI-08-0162
64. Dimopoulos MA, Germenis A, Savides P, Karayanis A, Fertakis A,
Dimopoulos C. Genetic markers in carcinoma of the prostate. Eur Urol.
(1984) 10:315–6. doi: 10.1159/000463818
65. Zhou L, Zhang X, Chen X, Liu L, Lu C, Tang X, et al. GC Glu416Asp
and Thr420Lys polymorphisms contribute to gastrointestinal cancer
susceptibility in a Chinese population. Int J Clin Exp Med. (2012) 5:72–9.
66. Poynter JN, Jacobs ET, Figueiredo JC, Lee WH, Conti DV, Campbell
PT, et al. Genetic variation in the vitamin D receptor (VDR) and
the vitamin D-binding protein (GC) and risk for colorectal cancer:
results from the Colon Cancer Family Registry. Cancer Epidemiol
Biomarkers Prev. (2010) 19:525–36. doi: 10.1158/1055-9965.EPI-
09-0662
67. McCullough ML, Stevens VL, Diver WR, Feigelson HS, Rodriguez C, Bostick
RM, et al. Vitamin D pathway gene polymorphisms, diet, and risk of
postmenopausal breast cancer: a nested case-control study. Breast Cancer
Res. (2007) 9:R9. doi: 10.1186/bcr1642
68. Ahn J, Albanes D, Berndt SI, Peters U, Chatterjee N, Freedman ND, et al.
Vitamin D-related genes, serum vitamin D concentrations and prostate
cancer risk. Carcinogenesis. (2009) 30:769–76. doi: 10.1093/carcin/bgp055
69. Martineau AR, Leandro AC, Anderson ST, Newton SM, Wilkinson KA,
Nicol MP, et al. Association between Gc genotype and susceptibility to
TB is dependent on vitamin D status. Eur Respir J. (2010) 35:1106–
12. doi: 10.1183/09031936.00087009
70. Karuwanarint P, Phonrat B, Tungtrongchitr A, Suriyaprom K,
Chuengsamarn S, Schweigert FJ, et al. Vitamin D-binding protein and
its polymorphisms as a predictor for metabolic syndrome. Biomark Med.
(2018) 12:465–73. doi: 10.2217/bmm-2018-0029
71. Xie CN, Yue M, Huang P, Tian T, Fan HZ, Wu MP, et al. Vitamin
D binding protein polymorphisms influence susceptibility to hepatitis C
virus infection in a high-risk Chinese population. Gene. (2018) 679:405–
11. doi: 10.1016/j.gene.2018.09.021
72. Karras SN, Koufakis T, Fakhoury H, Kotsa K. Deconvoluting the
biological roles of vitamin D-binding protein during pregnancy:
a both clinical and theoretical challenge. Front Endocrinol. (2018)
9:259. doi: 10.3389/fendo.2018.00259
73. Wang H, Cheng B, Chen Q, Wu S, Lv C, Xie G, et al. Time course of plasma
gelsolin concentrations during severe sepsis in critically ill surgical patients.
Crit Care. (2008) 12:R106. doi: 10.1186/cc6988
74. Dahl B, Schiodt FV, Ott P, Wians F, Lee WM, Balko J, et al.
Plasma concentration of Gc-globulin is associated with organ
dysfunction and sepsis after injury. Crit Care Med. (2003)
31:152–6. doi: 10.1097/00003246-200301000-00024
75. Kempker JA, Tangpricha V, Ziegler TR, Martin GS. Vitamin D
in sepsis: from basic science to clinical impact. Crit Care. (2012)
16:316. doi: 10.1186/cc11252
76. Gressner OA, Gao C, Siluschek M, Kim P, Gressner AM. Inverse
association between serum concentrations of actin-free vitamin D-binding
protein and the histopathological extent of fibrogenic liver disease or
hepatocellular carcinoma. Eur J Gastroenterol Hepatol. (2009) 21:990–
5. doi: 10.1097/MEG.0b013e3283293769
77. Schiodt FV, Ott P, Bondesen S, Tygstrup N. Reduced serum Gc-
globulin concentrations in patients with fulminant hepatic failure:
association with multiple organ failure. Crit Care Med. (1997) 25:1366–
70. doi: 10.1097/00003246-199708000-00025
78. Lind SE, Smith DB, Janmey PA, Stossel TP. Depression of gelsolin levels
and detection of gelsolin-actin complexes in plasma of patients with acute
lung injury. Am Rev Respir Dis. (1988) 138:429–34. doi: 10.1164/ajrccm/138.
2.429
79. Tannetta DS, Redman CW, Sargent IL. Investigation of the actin scavenging
system in pre-eclampsia. Eur J Obstetr Gynecol Reprod Biol. (2014) 172:32–
5. doi: 10.1016/j.ejogrb.2013.10.022
80. Speeckaert MM, Wehlou C, De Somer F, Speeckaert R, Van Nooten GJ,
Delanghe JR. Evolution of vitamin D binding protein concentration in sera
from cardiac surgery patients is determined by triglyceridemia. Clin Chem
Lab Med. (2010) 48:1345–50. doi: 10.1515/CCLM.2010.250
81. Dahl B, Schiodt FV, Gehrchen PM, Ramlau J, Kiaer T, Ott P. Gc-globulin is an
acute phase reactant and an indicator of muscle injury after spinal surgery.
Inflamm Res. (2001) 50:39–43. doi: 10.1007/s000110050722
Frontiers in Endocrinology | www.frontiersin.org 10 May 2019 | Volume 10 | Article 317
36
Bikle and Schwartz DBP and Free 25(OH)D
82. Koike K, Shinozawa Y, Yamazaki M, Endo T, Nomura R, Aiboshi J,
et al. Recombinant human interleukin-1alpha increases serum albumin, Gc-
globulin, and alpha1-antitrypsin levels in burned mice. Tohoku J Exp Med.
(2002) 198:23–9. doi: 10.1620/tjem.198.23
83. Meier U, Gressner O, Lammert F, Gressner AM. Gc-globulin:
roles in response to injury. Clin Chem. (2006) 52:1247–
53. doi: 10.1373/clinchem.2005.065680
84. Vasconcellos CA, Lind SE. Coordinated inhibition of actin-induced platelet
aggregation by plasma gelsolin and vitamin D-binding protein. Blood.
(1993) 82:3648–57.
85. Mc Leod JF, Kowalski MA, Haddad JG Jr. Interactions among serum vitamin
D binding protein, monomeric actin, profilin, and profilactin. J Biol Chem.
(1989) 264:1260–7.
86. Dueland S, Nenseter MS, Drevon CA. Uptake and degradation of
filamentous actin and vitamin D-binding protein in the rat. Biochem J. (1991)
274 (Pt 1):237–41. doi: 10.1042/bj2740237
87. Madden K, Feldman HA, Chun RF, Smith EM, Sullivan RM, Agan
AA, et al. Critically ill children have low vitamin D-binding protein,
influencing bioavailability of vitamin D. Ann Am Thorac Soc. (2015)
12:1654–61. doi: 10.1513/AnnalsATS.201503-160OC
88. Waldron JL, Ashby HL, Cornes MP, Bechervaise J, Razavi C, Thomas OL,
et al. Vitamin D: a negative acute phase reactant. J Clin Pathol. (2013)
66:620–2. doi: 10.1136/jclinpath-2012-201301
89. Leaf DE, Waikar SS, Wolf M, Cremers S, Bhan I, Stern L. Dysregulated
mineral metabolism in patients with acute kidney injury and risk of adverse
outcomes. Clin Endocrinol. (2013) 79:491–8. doi: 10.1111/cen.12172
90. Pihl TH, Jorgensen CS, Santoni-Rugiu E, Leifsson PS, Hansen EW, Laursen
I, et al. Safety pharmacology, toxicology and pharmacokinetic assessment
of human Gc globulin (vitamin D binding protein). Basic Clin Pharmacol
Toxicol. (2010) 107:853–60. doi: 10.1111/j.1742-7843.2010.00587.x
91. Gomme PT, Bertolini J. Therapeutic potential of vitamin D-binding
protein. Trends Biotechnol. (2004) 22:340–5. doi: 10.1016/j.tibtech.2004.
05.001
92. DiMartino SJ, Trujillo G, McVoy LA, Zhang J, Kew RR. Upregulation of
vitamin D binding protein (Gc-globulin) binding sites during neutrophil
activation from a latent reservoir in azurophil granules. Mol Immunol. (2007)
44:2370–7. doi: 10.1016/j.molimm.2006.10.011
93. Trujillo G, Habiel DM, Ge L, Ramadass M, Cooke NE, Kew RR. Neutrophil
recruitment to the lung in both C5a- and CXCL1-induced alveolitis is
impaired in vitamin D-binding protein-deficient mice. J Immunol. (2013)
191:848–56. doi: 10.4049/jimmunol.1202941
94. Shah AB, DiMartino SJ, Trujillo G, Kew RR. Selective inhibition
of the C5a chemotactic cofactor function of the vitamin D binding
protein by 1,25(OH)2 vitamin D3. Mol Immunol. (2006) 43:1109–
15. doi: 10.1016/j.molimm.2005.07.023
95. Swamy N, Ray R. Fatty acid-binding site environments of serum vitamin
D-binding protein and albumin are different. Bioorg Chem. (2008) 36:165–
8. doi: 10.1016/j.bioorg.2008.02.002
96. Ena JM, Esteban C, Perez MD, Uriel J, Calvo M. Fatty acids bound to
vitamin D-binding protein (DBP) from human and bovine sera. Biochem Int.
(1989) 19:1–7.
97. Yamamoto N, Homma S. Vitamin D3 binding protein (group-specific
component) is a precursor for the macrophage-activating signal factor
from lysophosphatidylcholine-treated lymphocytes. Proc Natl Acad Sci USA.
(1991) 88:8539–43. doi: 10.1073/pnas.88.19.8539
98. Yamamoto N, Homma S, Millman I. Identification of the serum factor
required for in vitro activation of macrophages. Role of vitamin D3-binding
protein (group specific component, Gc) in lysophospholipid activation of
mouse peritoneal macrophages. J Immunol. (1991) 147:273–80.
99. Swamy N, Ghosh S, Schneider GB, Ray R. Baculovirus-expressed vitamin
D-binding protein-macrophage activating factor (DBP-maf) activates
osteoclasts and binding of 25-hydroxyvitamin D(3) does not influence
this activity. J Cell Biochem. (2001) 81:535–46. doi: 10.1002/1097-
4644(20010601)81:3<535::aid-jcb1067>3.0.co;2-6
100. Schneider GB, Benis KA, Flay NW, Ireland RA, Popoff SN. Effects of vitamin
D binding protein-macrophage activating factor (DBP-MAF) infusion on
bone resorption in two osteopetrotic mutations. Bone. (1995) 16:657–
62. doi: 10.1016/8756-3282(95)00118-W
101. Korbelik M, Naraparaju VR, Yamamoto N. Macrophage-directed
immunotherapy as adjuvant to photodynamic therapy of cancer. Br J
Cancer. (1997) 75:202–7. doi: 10.1038/bjc.1997.34
102. Koga Y, Naraparaju VR, Yamamoto N. Antitumor effect of vitamin
D-binding protein-derived macrophage activating factor on Ehrlich
ascites tumor-bearing mice. Proc Soc Exp Biol Med. (1999) 220:20–
6. doi: 10.1046/j.1525-1373.1999.d01-3.x
103. Kisker O, Onizuka S, Becker CM, Fannon M, Flynn E, D’Amato R,
et al. Vitamin D binding protein-macrophage activating factor (DBP-maf)
inhibits angiogenesis and tumor growth in mice. Neoplasia. (2003) 5:32–
40. doi: 10.1016/S1476-5586(03)80015-5
104. Yamamoto N, Naraparaju VR, Asbell SO. Deglycosylation of serum vitamin
D3-binding protein leads to immunosuppression in cancer patients. Cancer
Res. (1996) 56:2827–31.
105. Yamamoto N, Naraparaju VR, Urade M. Prognostic utility of serum
alpha-N-acetylgalactosaminidase and immunosuppression resulted from
deglycosylation of serum Gc protein in oral cancer patients. Cancer Res.
(1997) 57:295–9.
106. Recant L, Riggs DS. Thyroid function in nephrosis. J Clin Invest. (1952)
31:789–97. doi: 10.1172/JCI102664
107. Refetoff S. Thyroid hormone serum transport proteins. In: De Groot LJ,
Chrousos G, Dungan K, Feingold KR, Grossman A, Hershman JM, et al.,
editors. Endotext, South Dartmouth, MA: MD text.com (2000).
108. Siiteri PK, Murai JT, Hammond GL, Nisker JA, Raymoure WJ, Kuhn RW.
The serum transport of steroid hormones. Recent Prog Horm Res. (1982)
38:457–510. doi: 10.1016/B978-0-12-571138-8.50016-0
109. Bikle DD, Gee E, Halloran B, Haddad JG. Free 1,25-dihydroxyvitamin D
levels in serum from normal subjects, pregnant subjects, and subjects with
liver disease. J Clin Invest. (1984) 74:1966–71. doi: 10.1172/JCI111617
110. Bikle DD, Halloran BP, Gee E, Ryzen E, Haddad JG. Free 25-
hydroxyvitamin D levels are normal in subjects with liver disease and
reduced total 25-hydroxyvitamin D levels. J Clin Invest. (1986) 78:748–
52. doi: 10.1172/JCI112636
111. Bikle D, Bouillon R, Thadhani R, Schoenmakers I. Vitamin D
metabolites in captivity? Should we measure free or total 25(OH)D
to assess vitamin D status? J Steroid Biochem Mol Biol. (2017)
173:105–16. doi: 10.1016/j.jsbmb.2017.01.007
112. Schwartz JB, Lai J, Lizaola B, Kane L, Weyland P, Terrault NA, et al.
Variability in free 25(OH) vitamin D levels in clinical populations. J Steroid
Biochem Mol Biol. (2014) 144 (Pt A):156–8. doi: 10.1016/j.jsbmb.2013.11.006
113. Bikle DD, Siiteri PK, Ryzen E, Haddad JG. Serum protein binding
of 1,25-dihydroxyvitamin D: a reevaluation by direct measurement
of free metabolite levels. J Clin Endocrinol Metab. (1985) 61:969–
75. doi: 10.1210/jcem-61-5-969
114. 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)
63:954–9. doi: 10.1210/jcem-63-4-954
115. Schwartz JB, Lai J, Lizaola B, Kane L, Markova S, Weyland P, et al.
A comparison of measured and calculated free 25(OH) vitamin D
levels in clinical populations. J Clin Endocrinol Metab. (2014) 99:1631–
7. doi: 10.1210/jc.2013-3874
116. Muller MJ, Volmer DA. Mass spectrometric profiling of vitamin D
metabolites beyond 25-hydroxyvitamin D. Clin Chem. (2015) 61:1033–
48. doi: 10.1373/clinchem.2015.241430
117. Binkley N, Carter GD. Toward clarity in clinical vitamin D status assessment:
25(OH)D assay standardization. Endocrinol Metab Clin North Am. (2017)
46:885–99. doi: 10.1016/j.ecl.2017.07.012
118. Kilpatrick LE, Phinney KW. Quantification of total vitamin-D-binding
protein and the glycosylated isoforms by liquid chromatography-
isotope dilution mass spectrometry. J Proteome Res. (2017)
16:4185–95. doi: 10.1021/acs.jproteome.7b00560
119. Nielson CM, Jones KS, Bouillon R, Osteoporotic Fractures in Men Research
G, Chun RF, Jacobs J, et al. Role of assay type in determining free 25-
hydroxyvitamin D levels in diverse populations. N Engl J Med. (2016)
374:1695–6. doi: 10.1056/NEJMc1513502
120. Nielson CM, Jones KS, Chun RF, Jacobs JM, Wang Y, Hewison M, et al.
Free 25-hydroxyvitamin D: impact of vitamin D binding protein assays on
Frontiers in Endocrinology | www.frontiersin.org 11 May 2019 | Volume 10 | Article 317
37
Bikle and Schwartz DBP and Free 25(OH)D
racial-genotypic associations. J Clin Endocrinol Metab. (2016) 101:2226–
34. doi: 10.1210/jc.2016-1104
121. Lee MJ, Kearns MD, Smith EM, Hao L, Ziegler TR, Alvarez JA, et al. Free
25-hydroxyvitamin D concentrations in cystic fibrosis. Am J Med Sci. (2015)
350:374–9. doi: 10.1097/MAJ.0000000000000592
122. Sollid ST, Hutchinson MY, Berg V, Fuskevag OM, Figenschau Y, Thorsby
PM, et al. Effects of vitamin D binding protein phenotypes and vitamin
D supplementation on serum total 25(OH)D and directly measured free
25(OH)D. Eur J Endocrinol. (2016) 174:445–52. doi: 10.1530/EJE-15-1089
123. Jassil NK, Sharma A, Bikle D, Wang X. Vitamin D binding protein and 25-
hydroxyvitamin D levels: emerging clinical applications. Endocrine Pract.
(2017) 23:605–13. doi: 10.4158/EP161604.RA
124. Tsuprykov O, Buse C, Skoblo R, Haq A, Hocher B. Reference intervals for
measured and calculated free 25-hydroxyvitamin D in normal pregnancy. J
Steroid Biochem Mol Biol. (2018) 181:80–7. doi: 10.1016/j.jsbmb.2018.03.005
125. van Hoof HJ, de Sevaux RG, van Baelen H, Swinkels LM, Klipping C,
Ross HA, et al. Relationship between free and total 1,25-dihydroxyvitamin
D in conditions of modified binding. Eur J Endocrinol. (2001) 144:391–
6. doi: 10.1530/eje.0.1440391
126. Walsh JS, Evans AL, Bowles S, Naylor KE, Jones KS, Schoenmakers I,
et al. Free 25-hydroxyvitamin D is low in obesity, but there are no
adverse associations with bone health. Am J Clin Nutr. (2016) 103:1465–
71. doi: 10.3945/ajcn.115.120139
127. Powe CE, Evans MK, Wenger J, Zonderman AB, Berg AH, Nalls
M, et al. Vitamin D-binding protein and vitamin D status of black
Americans and white Americans. N Engl J Med. (2013) 369:1991–
2000. doi: 10.1056/NEJMoa1306357
128. Schwartz JB, Kane L, Bikle D. Response of vitamin D concentration
to vitamin D3 administration in older adults without sun exposure:
a randomized double-blind trial. J Am Geriatr Soc. (2016) 64:65–
72. doi: 10.1111/jgs.13774
129. Srikanth P, Chun RF, Hewison M, Adams JS, Bouillon R, Vanderschueren
D, et al. Associations of total and free 25OHD and 1,25(OH)2D with serum
markers of inflammation in older men. Osteoporosis Int. (2016) 27:2291–
300. doi: 10.1007/s00198-016-3537-3
130. Shieh A, Chun RF, Ma C, Witzel S, Meyer B, Rafison B, et al. Effects of
high-dose vitamin D2 versus D3 on total and free 25-hydroxyvitamin D
and markers of calcium balance. J Clin Endocrinol Metab. (2016) 101:3070–
8. doi: 10.1210/jc.2016-1871
131. Shieh A, Ma C, Chun RF, Wittwer-Schegg J, Swinkels L, Huijs T, et al.
Associations between change in total and free 25-hydroxyvitamin D with
24,25-dihydroxyvitamin D and parathyroid hormone. J Clin Endocrinol
Metab. (2018) 103:3368–75. doi: 10.1210/jc.2018-00515
Conflict of Interest Statement: The authors declare that the research was
conducted in the absence of any commercial or financial relationships that could
be construed as a potential conflict of interest.
Copyright © 2019 Bikle and Schwartz. This is an open-access article distributed
under the terms of the Creative Commons Attribution License (CC BY). The use,
distribution or reproduction in other forums is permitted, provided the original
author(s) and the copyright owner(s) are credited and that the original publication
in this journal is cited, in accordance with accepted academic practice. No use,
distribution or reproduction is permitted which does not comply with these terms.
Frontiers in Endocrinology | www.frontiersin.org 12 May 2019 | Volume 10 | Article 317
38
REVIEW
published: 10 July 2019
doi: 10.3389/fendo.2019.00470
Frontiers in Endocrinology | www.frontiersin.org 1July 2019 | Volume 10 | Article 470
Edited by:
Zhongjian Xie,
Central South University, China
Reviewed by:
Giovanni Lombardi,
Istituto Ortopedico Galeazzi
(IRCCS), Italy
Stefano Pagano,
University of Perugia, Italy
*Correspondence:
Richard R. Kew
richard.kew@stonybrookmedicine.edu
Specialty section:
This article was submitted to
Bone Research,
a section of the journal
Frontiers in Endocrinology
Received: 30 April 2019
Accepted: 28 June 2019
Published: 10 July 2019
Citation:
Kew RR (2019) The Vitamin D Binding
Protein and Inflammatory Injury: A
Mediator or Sentinel of Tissue
Damage? Front. Endocrinol. 10:470.
doi: 10.3389/fendo.2019.00470
The Vitamin D Binding Protein and
Inflammatory Injury: A Mediator or
Sentinel of Tissue Damage?
Richard R. Kew*
Department of Pathology, Stony Brook Cancer Center, Renaissance School of Medicine, Stony Brook University,
Stony Brook, NY, United States
Neutrophils are the most abundant type of white blood cell in most mammals including
humans. The primary role of these cells is host defense against microbes and clearance of
tissue debris in order to facilitate wound healing and tissue regeneration. The recruitment
of neutrophils from blood into tissues is a key step in this process and is mediated by
numerous different chemoattractants. The neutrophil migratory response is essential for
host defense and survival, but excessive tissue accumulation of neutrophils is observed in
many inflammatory disorders and strongly correlates with disease pathology. The vitamin
D binding protein (DBP) is a circulating multifunctional plasma protein that can significantly
enhance the chemotactic activity of neutrophil chemoattractants both in vitro and in vivo.
Recent in vivo studies using DBP deficient mice showed that DBP plays a larger and
more central role during inflammation since it induces selective recruitment of neutrophils,
and this cofactor function is not restricted to C5a, as prior in vitro studies indicated,
but can enhance chemotaxis to many chemoattractants. DBP also is an extracellular
scavenger for actin released from damaged/dead cells and formation of DBP-actin
complexes is an immediate host response to tissue injury. Recent in vitro evidence
indicates that DBP bound to G-actin, and not free DBP, functions as an indirect but
essential cofactor for neutrophil migration. DBP-actin complexes always will be formed
regardless of what initiated an inflammation, since release of actin from damaged cells
is a common feature in all types of injury and DBP is abundant and ubiquitous in all
extracellular fluids. Indeed, these complexes have been detected in blood and tissue
fluids from both humans and experimental animals following various forms of injury.
The published data strongly supports the premise that DBP-actin complexes are the
functional neutrophil chemotactic cofactor that enhances neutrophil chemotaxis in vitro
and augments neutrophilic inflammation in vivo. This review will assess the fundamental
role of DBP in neutrophilic inflammation and injury.
Keywords: inflammation, neutrophil accumulation, chemotactic factor, vitamin D binding protein (DBP), tissue
injury
39
Kew DBP and Inflammatory Injury
HISTORICAL CONTEXT AND BRIEF
BACKGROUND
The vitamin D binding protein (DBP, also abbreviated VDBP)
was initially named the group-specific component of serum
(abbreviated as Gc-globulin). Jan Hirschfeld is credited with the
first publication in 1959 describing the migration pattern of an
unknown serum protein using agarose gel electrophoresis and
rabbit anti-human serum (i.e., immunoelectrophoresis) (1). This
report described qualitative differences in the electrophoretic
migration of α2-globulin proteins in serum obtained from
10 normal healthy blood donors (1). These immunoreactive
proteins would display a consistent migration pattern (either
fast, intermediate, or slow) that was specific to an individual
donor, and thus was named the group-specific component of
serum (1). This paper, together with two subsequent papers by
Hirschfeld published the following year (2,3), initiated this six
decade long investigation into the structure and function of DBP.
A major milestone for DBP research occurred in 1975 when
Diager et. al. reported that the serum group-specific component
binds vitamin D, and consequently, the authors proposed to
name the protein the vitamin D binding α-globulin (4), later
shortened to the vitamin D binding protein. A few years later
several reports described what initially was thought to be a
larger cellular form of DBP (57) but subsequently was shown
unequivocally to be plasma DBP binding to G-actin monomers in
a 1:1 molar complex (8). Thus, by 1980, DBP was known to have
two distinct binding functions: vitamin D and G-actin. Structural
studies would later confirm the vitamin D sterol and G-actin
binding pockets in DBP (9,10). Beginning in the mid-1980’s
and continuing into the early 1990’s, numerous reports identified
DBP and plasma gelsolin as part of the actin scavenging system
in blood. These proteins work in tandem to clear extracellular
actin released from dead or damaged cells for removal from
the circulation, largely in the liver (11). Furthermore, several
studies demonstrated that serious injury (burns, traumatic injury,
acetaminophen-induced hepatotoxicity) generate large amounts
of extracellular actin that consumes DBP and reduces its plasma
concentration (1215). Low levels of plasma DBP directly
correlate with poor overall survival in these studies (1215).
Although the actin binding properties of DBP have been clearly
established and shown to be physiologically relevant, this role
largely has been overshadowed by the vitamin D sterol binding
function, particularly over the past 15 years with increasing
awareness of the essential role of vitamin D in health and disease.
Besides, the vitamin D binding protein is not the best choice
of name to promote its actin binding function! There are many
facets of this multifunctional plasma protein, several of which are
discussed in other reviews of this series. The topic herein will
focus primarily on the actin binding capacity of DBP, how this
function plays a role in chemotaxis enhancement of neutrophils
and may act as a possible mediator of inflammatory tissue injury.
DBP-ACTIN COMPLEXES AND TISSUE
INJURY
Tissue injury often causes cell damage and death leading to
the release of intracellular contents into extracellular fluids.
Many intracellular molecules (HMGB1, mitochondrial DNA,
ATP, etc.) have different roles in the extracellular environment
and function as “alarmins” (also known as danger-associated
molecular patterns or DAMPs) to signal the immune system
to the presence of tissue injury (16). Although numerous
alarmins have been described and their role in tissue injury
well-characterized, it is quite surprising that very little attention
has been paid to actin, the major cytoskeletal protein that
is the most abundant intracellular protein in any organism.
Actin essentially exists in two states inside a cell: monomeric
globular actin (G-actin), or G-actin polymerized into filaments
(F-actin) (17). These two forms are in a constant state of
polymerization and depolymerization that is highly regulated by
several intracellular actin binding proteins, and this dynamic
process is very prevalent in motile cells such as leukocytes
(18). During tissue injury large quantities of actin can be
released into extracellular fluids where the protein escapes
normal intracellular regulatory mechanisms and the protein
spontaneously will form F-actin filaments. In animal models,
extracellular F-actin filaments have been shown to alter the
coagulation and fibrinolytic systems leading to occlusion and
damage of the microcirculation (particularly in the lung) in a
manner similar to how fibrin damages the microvasculature in
disseminated intravascular coagulation (15,19). Accordingly, an
effective extracellular scavenging system has evolved to dispose
of actin released from dead or damaged cells (15). The system
is composed of two plasma proteins that have complementary
functions: gelsolin, which severs F-actin filaments into G-actin
monomers, and DBP that binds G-actin in a high affinity (Kd
of 109M) 1:1 molar complex for transport and eventual
clearance primarily in the liver. This process consumes DBP and
a decreased plasma concentration of actin-free DBP has been
shown to be an effective but indirect marker of tissue injury in
cases of severe trauma in both humans and animal models (15).
Although there has been interest in the potential therapeutic role
of gelsolin as an extracellular actin scavenger to treat several
conditions, less attention has been focused on its partner DBP,
despite the fact that over the past 30 years numerous studies
(>400 listed on PubMed) have reported that DBP is associated
with various acute and chronic diseases in several different organs
[reviewed in (20,21)]. There is no apparent common connection
in these various studies, but a very broad view of all these reports
would suggest that DBP has a fundamental role during tissue
injury, perhaps due to its actin scavenging function.
The vitamin D binding protein is an abundant plasma
protein that is part of the albumin gene family and shares
considerable amino acid homology and structural similarity
with these proteins (11). Several groups have published the
crystal structure of DBP that revealed an α-helix triple domain
arrangement (characteristic of the albumin family) that forms a
broad U-shaped or saddle-shaped molecule (9,22,23). The N-
terminal (domain I) and the C-terminal domains (III) form the
front and back of the saddle and domain II the seat (10,22,23).
This shape is designed to perfectly fit a molecule of G-actin,
and this has been confirmed by crystal structure analysis of
DBP bound to G-actin (10). Vitamin D sterol binding pocket
in domain I is distinct from the actin binding domain, and a
molecule of DBP can bind both ligands simultaneously without
Frontiers in Endocrinology | www.frontiersin.org 2July 2019 | Volume 10 | Article 470
40
Kew DBP and Inflammatory Injury
apparent alteration in binding affinity or protein function (24).
Although DBP mRNA expression has been reported in many
different tissues, plasma DBP is synthesized by hepatocytes in
the liver and circulates in blood with a range of 5–9 µM (300–
500 µg/ml) (11). It has a relatively rapid turnover in plasma with
a half-life of about 2 days (compared to 22 days for albumin)
(25). In contrast to albumin whose plasma levels decrease during
inflammation, DBP levels in blood are stable or rise slightly
during the acute phase response of inflammation (26). Moreover,
DBP is ubiquitous in vivo and significant quantities (0.1–1 µM
range) have been detected in all extracellular fluid compartments
(cerebrospinal, bronchoalveolar, synovial, etc.) (25,26). Although
DBP is the primary transport molecule for vitamin D sterols in
the blood and extracellular fluids, its concentration in plasma
is much higher than vitamin D. Normally only 1–2% of the
total circulating DBP pool has vitamin D bound and this
percentage never rises above 5% (27). This is in contrast with
other transport proteins in blood where about 50% of the total
protein pool is bound with ligand (25). This fact prompted
many to speculate that DBP must have other essential functions,
perhaps related to its ability to scavenge G-actin (28). Indeed,
significant tissue injury can result in a large percentage of
circulating DBP complexed with actin (1215). Actin-induced
depletion of plasma DBP to levels below 3.5 µM (200 µg/ml)
have been shown to be an effective but indirect marker of tissue
injury that correlates with poor prognosis in cases of sepsis,
multiple trauma and acetaminophen-induced liver failure (12
15). Clinical outcome and decreased plasma levels of DBP have a
statistical correlation similar to the other outcome metrics such
as the APACHE II score (sepsis), Kings College criteria (liver
failure) and the TRISS-like method (multiple trauma) (1315).
The vitamin D binding protein and actin are abundant
and ubiquitous proteins of the intra- and extracellular
compartments, their expression is stable during inflammation
and cell injury causes immediate complex formation. Thus,
global transcriptome analysis of mRNAs increased or decreased
during inflammatory injury most likely would not identify
actin or DBP (29). For these reasons we believe that the
potential role of DBP-actin complexes during inflammation has
been overlooked. Varying levels of DBP-actin complexes are
continually formed in vivo as a result of minor tissue trauma,
menstrual cycles, infections, surgery, etc. Hence, low levels of
circulating DBP-actin would be a routine physiological process
and should be transient and inconsequential. On the other hand,
prolonged generation and/or high concentrations of DBP-actin
in extracellular fluids potentially could act as a danger signal
(alarmin) of ongoing and significant tissue injury. Although
previous research has focused on actin-free DBP in plasma,
the role of DBP-actin complexes in tissue injury has not been
determined, most likely because it has been assumed that these
complexes are inactive by-products of cell damage that are
rapidly cleared from the circulation. Interestingly, a previous
study examining fulminant hepatic necrosis (FHN) in humans
showed that 72% of plasma DBP was complexed with actin in
patients who died of the disease whereas only 22% of total DBP
was bound to actin in FHN survivors (12). However, while this
data was very compelling, the authors of this study focused on
actin-free DBP, not the level of DBP-actin complexes. Although
DBP-actin complexes generally have been viewed as benign
by-products of cell injury, recent studies from our lab (both in
vitro and in vivo) have shown that these complexes may serve
as an alarmin and possess a cytokine-like function. Thus, the
accumulated evidence has shown that DBP, via its actin binding
function, likely plays a role in both the mediation and resolution
of tissue injury.
CELL-ASSOCIATED DBP
The vitamin D binding protein interacts with many different
cell types to achieve its diverse functions (delivery of vitamin D
sterols, binding and/or clearance of G-actin, chemotactic cofactor
function, macrophage activating-factor), and it appears that the
protein must first bind to its target cell surface in order to mediate
these effects. Numerous studies have reported DBP on the
surface of most cell types including B-lymphocytes (3032), T-
lymphocytes (33), testicular cells (34), placental cytotrophoblasts
(35,36), pancreatic acinar cells (37), monocytes (38), neutrophils
(39), and renal proximal tubule cells (40). It is clear that cell-
associated DBP is not a novel cellular form but rather circulating
DBP bound to the cell surface (41). Although binding to the
plasma membrane is the first step in the interaction of DBP
with cells, it is unlikely that a cell surface binding site for
DBP would be a specific high affinity (Kd108M) receptor.
DBP is abundant and ubiquitous in all fluid compartments
and circulating blood leukocytes are bathed in 5–9 µM DBP
(11). Therefore, a relatively low affinity binding site with a Kd
significantly above the plasma concentration of DBP would seem
most logical, otherwise leukocytes in blood could act as a “sink”
for DBP and diminish the circulating pool. Several studies have
shown that DBP binds with low affinity to megalin, also known
as LDL receptor related protein 2 (LRP2) (4244). Megalin is a
large scavenger receptor that is primarily expressed on the surface
of epithelial cells in the kidney (proximal tubules), endocrine
glands, and reproductive organs (45). Megalin on renal proximal
tubules binds and captures DBP (both free and bound to vitamin
D) in glomerular filtrate for re-uptake into the circulation (45).
DBP also binds specifically, but with low avidity, to chondroitin
sulfate glycosaminoglycans (CS-GSGs), and more specifically to
the CS-GAG group on CD44, which is widely expressed on
most leukocytes (46,47). However, several papers have reported
that DBP bound to the surface of cells cannot be removed
by numerous high salt and/or detergent washes, suggesting a
relatively tight binding site (30,33,38).
Our lab has extensively investigated the binding of DBP to
human neutrophils and myeloid cell lines (U937, HL-60) since
cell binding is the critical first step in the process of DBP
functioning as a chemotactic cofactor. DBP does not follow the
kinetics of saturable binding to a single high-affinity receptor,
but instead displays a multiphasic, time-dependent interaction
with neutrophils over the course of 60 min at 37C: weak
binding (5–20 min), strong binding (20–50 min), shedding into
the extracellular fluid (>45 min) (46). There is very little (if any)
binding of radiolabeled DBP to cells incubated in an ice bath
Frontiers in Endocrinology | www.frontiersin.org 3July 2019 | Volume 10 | Article 470
41
Kew DBP and Inflammatory Injury
(1C), but at 37C three distinct phases of DBP binding are
observed, indicating that a dynamic process is required to express
this binding site. In addition, if neutrophils are first stimulated
with chemotactic factors or calcium ionophores at 37C prior to
incubation with labeled DBP, the delay in tight binding phase
is essentially eliminated. The initial weak binding of DBP to
neutrophils probably is mediated by low avidity molecules such
as CD44 or other chondroitin sulfate proteoglycans upregulated
from internal stores in neutrophil cytoplasmic granules (46,48).
These weak binding molecules may serve to capture DBP from
extracellular fluids and loosely tether it to the cell surface.
The biochemical characterization of the tight binding site
for DBP on the plasma membrane of neutrophils, and
mechanisms of the subsequent protease-mediated shedding,
were very extensive (46,4850) and showed that: (a) DBP
bound to the plasma membrane can only be dissociated
using harsh denaturing conditions; (b) solubilization of cells
revealed that the bound DBP partitions to the detergent
insoluble fraction, which contains the actin cytoskeleton; (c)
confocal immunofluorescence microscopy revealed that DBP
and actin co-localize on the surface of neutrophils; (d)
immunoprecipitation of DBP bound to (or shed from) cells,
followed by mass spectrometry analysis showed that the major
binding partners were actin, CD44 and annexin A2. Annexin
A2 is known as a molecular facilitator since it binds multiple
diverse molecules (including CD44 and actin) to assemble plasma
membrane complexes in a phospholipid and calcium-dependent
manner (51). Interestingly, two reports published more than a
dozen years before our studies on neutrophil cell surface DBP
binding site, described that DBP was tightly bound to actin on
the surface of human B lymphocytes (30) and monocytes (38).
The third phase, DBP shedding from the cell surface, was
shown to be due to the action of the enzyme elastase on the
DBP binding site, since DBP is not modified (cleaved) during
this process (49). Elastase is a physiologically important protease
with broad substrate specificity, its expression is restricted mainly
to neutrophils and abundant quantities of this protease are
stored in the azurophil (primary) granules in the cytoplasm
of both immature and mature neutrophils (52). Inhibition of
elastase will inhibit shedding and cause DBP to accumulate on
the plasma membrane, and also will inhibit the chemotactic
cofactor function of DBP (49). Previous reports have described
proteolytically active elastase on the cell surface of neutrophils
in vitro (53,54). Interestingly, both DBP binding, and shedding
of the binding site, are constitutive processes and occur in the
absence of an inflammatory or chemotactic stimulus, however
these stimuli accelerate the process (46,49). Moreover, as will be
discussed below, DBP binding to cell surface actin on neutrophils
and subsequent elastase-mediated shedding from the plasma
membrane, may be a key step in understanding its function as
a chemotactic cofactor.
In addition to DBP uptake from fluids, we also have previously
reported that human neutrophils contain an intracellular store
of DBP in specific (secondary) granules (39), and this finding
was verified in a follow-up study investigating the DBP binding
site in neutrophils (46). Neutrophil specific granules contain a
diverse array of molecules including several binding proteins
such as haptoglobin and vitamin B12 binding protein (55). It is
not clear why these cells have DBP stored in intracellular granules
formed during myelocyte stage of neutrophil development in
the bone marrow (56). Perhaps cells utilize this store of DBP
during chemotaxis and phagocytosis when there is dynamic
rearrangement of cell structures and release of granule contents
(56). The amount of DBP was calculated to be 3 ng/106
neutrophils, a rather small quantity but considering that an
inflammatory exudate may contain billions of neutrophils, the
amount of DBP that neutrophils potentially could release in an
inflammatory lesion may not be insignificant.
NEUTROPHILS, INFLAMMATORY INJURY,
AND THE CHEMOTACTIC COFACTOR
FUNCTION OF DBP
Neutrophils are the primary “rapid response” cells of the innate
immune system that are essential for host defense (57,58).
Individuals with a marked reduction in circulating neutrophils
(neutropenia), either due to a rare congenital abnormality
or more commonly a consequence of cancer chemotherapy,
are highly susceptible to severe and often life-threatening
bacterial and fungal infections. The importance of maintaining
adequate numbers of circulating neutrophils is highlighted by
the fact that 60% of the total bone marrow output of all
blood cells is dedicated just to produce neutrophils (59). It is
estimated that the average healthy person has a steady-state
production of 100 billion neutrophils per day under homeostatic
conditions, whereas cell production increases significantly and
rapidly during an infection, a process known as emergency
granulopoiesis (60,61). However, it has been well-known
for more than a century that excessive tissue accumulation
of neutrophils is observed in many inflammatory disorders
(neutrophilic inflammation). Cytotoxic products from activated
neutrophils mediate significant tissue injury and are linked to
the pathogenesis of numerous acute and chronic diseases (57
59). The destructive potential wrought by excessive numbers of
responding neutrophils is highlighted by the fact that this cell
type can liquify and obliterate tissue, i.e., a cavity resulting from
an abscess (62). There has been remarkable progress over the
past 5–8 years in understanding various aspects of neutrophil
biology, from their birth in the bone marrow to their death at
sites of inflammation and everything in between. Perhaps most
interesting is that neutrophils possess considerable plasticity and
are far more adaptable to their environment than previously
thought. Furthermore, neutrophils can display several disparate
functions that contradict their long-time moniker as “masters of
tissue destruction.” Many excellent reviews on various aspects of
neutrophil biology have been published recently (6366).
Migration of neutrophils from the bloodstream into various
tissues is a critical stage during inflammation triggered by both
infectious and non-infectious stimuli (63). Although much is
known about the chemoattractants and their receptors that
initiate and direct the neutrophil migratory response, little
is known about factors in physiological fluids that regulate
tissue recruitment of these cells. More than 35 years ago
Frontiers in Endocrinology | www.frontiersin.org 4July 2019 | Volume 10 | Article 470
42
Kew DBP and Inflammatory Injury
several investigators had demonstrated that normal human
serum possesses a heat stable chemotactic enhancing activity for
complement activation peptide C5a (6769). A 60 kDa protein
(called co-chemotaxin) was partially purified from human serum
and shown to be capable of enhancing the neutrophil chemotactic
activity of C5a and its stable degradation product C5a des
Arg (70). Previously, we were the first of several groups to
identify that DBP was the serum co-chemotaxin for C5a (71,
72), and several other groups subsequently confirmed our
initial observations that DBP can augment the chemotactic
activity of C5a/C5a des Arg (7379). These studies all reported
that DBP does not possess chemotactic activity but requires
chemoattractants to exert its cofactor activity. Other studies
also reported that DBP is not able to augment other C5a-
mediated functions in neutrophils such as oxidant generation
and degranulation (release of cytoplasmic granule contents)
(71,79). But most curious was the observation that DBP
could not enhance the chemotactic activity of other major
neutrophil chemoattractants: formylated peptides, CXCL8 (IL-
8), leukotriene B4or platelet activating factor (71,72), leading to
the general consensus that the chemotactic enhancing properties
of DBP appeared to be restricted to C5a/C5a des Arg. All of
these reports used an in vitro chemotaxis assay, employing
either blind-well or Boyden chambers, to measure an increase
in neutrophil migration when DBP was added. Although this
assay is very sensitive and quantitative, it has limitations that
resulted in the mistaken conclusion that DBP was specific
for complement peptide C5a. C5a is a very potent neutrophil
chemotactic factor and on a molar basis is 10–50 times more
potent than other major chemoattractants when tested in vitro
(71,80). Therefore, the chemotactic enhancing activity of DBP
during the short incubation (30 min) filter-based assays was
particularly noticeable when C5a was used as the stimulus.
However, utilizing a different in vitro assay (under agarose), that
requires a longer incubation period (4 h), revealed that DBP
could enhance neutrophil movement to other chemoattractants
as well (81). Moreover, in vivo studies using the DBP null mice
(discussed below) have shown that the chemotactic cofactor
activity of DBP is not specific for C5a as previously thought
but can augment the chemotactic activity of perhaps many
leukocyte chemoattractants. These recent in vitro and in vivo
studies described below have helped to better define how DBP
functions to enhance neutrophil chemotaxis that may contribute
to neutrophilic inflammation.
LESSONS FROM THE DBP NULL MOUSE
The initial reports describing the chemotactic cofactor function
of DBP were received enthusiastically at that time in the
inflammation research community (71,72). However, that high
level of interest faded with the subsequent failure to describe
a clear mechanism of chemotaxis enhancement, and to provide
confirmation that DBP enhances neutrophil chemotaxis in
vivo. Previous attempts to define a mechanism using in vitro
approaches were not successful. For example, our lab performed
numerous chemical cross-linking and co-immunoprecipitation
experiments that demonstrated clearly DBP does not physically-
associate with the C5a receptor (50). In addition, we reported
that and DBP does not bind to C5a, and cell binding of C5a and
DBP are independent events (82). Other groups have shown that
DBP does not alter the number of neutrophil C5a receptors or
the receptor Kdfor C5a, thereby discounting another obvious
explanation for its co-chemotactic effect (79,83). The question
of physiological relevance and whether DBP enhances neutrophil
recruitment to tissues in vivo needed to wait more than 10 years
until a DBP null mouse was generated.
The vitamin D binding protein has been very well-studied in
multiple different populations worldwide for over 50 years, but
no natural homozygous deficiency of DBP had been reported in
humans, or to the best of our knowledge, any mammal. There was
no clear reason why a DBP-deficiency was never identified but
it was widely speculated that a DBP deletion may be embryonic
lethal. Nevertheless, Nancy Cooke’s lab at the University of
Pennsylvania produced the first mice that were homozygous
null for DBP (84). These mice were healthy, of similar size
and appearance as wild-type mice, and the DBP null males and
females were fertile and produced normal sized litters (84). This
phenotype was somewhat surprising at the time given the fact
that a natural DBP deficiency had not been observed. Analysis
of blood chemistry values revealed that DBP null mice had
essentially the same levels of serum calcium, phosphorus, PTH,
and alkaline phosphatase as DBP sufficient wild type mice when
fed a standard vitamin D replete mouse chow diet (84). However,
the serum of DBP null mice had >95% reduction in both 25(OH)
and 1,25 (OH)2vitamin D (84). Moreover, when placed on a
vitamin D deficient diet, the DBP null mice quickly developed
secondary hyperparathyroidism and bone mineralization defects
such as osteoid thickening that were not seen in DBP sufficient
wild-type mice (84). The initial DBP null strain was backcrossed
onto a C57BL/6J background for 10 generations and was used
in further studies which showed that the lack of circulating
DBP does not alter the tissue distribution, uptake, activation or
biological potency of vitamin D (85). Thus, DBP does not alter
bioavailable vitamin D but appears to function as a circulating
reservoir of 25(OH) vitamin D.
Since no case of homozygous deficiency in humans has been
reported, a long-standing unresolved question has been: can DBP
null mice function as a murine model for DBP deficiency in
humans? Surprisingly, during the preparation of this review,
the first report of a DBP deficient human was published
(86). The individual is a 58 year-old female who presented
with long-standing and progressive ankylosing spondylitis and
severe vitamin D deficiency that did not respond to vitamin
D supplementation. Laboratory results showed a deletion of
the DBP (GC) gene and corresponding absence of circulating
DBP. Although this individual had a profound deficiency in
both 25(OH) and 1,25 (OH)2vitamin D, her blood calcium
levels were normal (86). The bone abnormalities and blood
chemistry values of this DBP deficient woman were similar to
those observed in DBP null mice fed a vitamin D-deficient diet
(84,86). In vitro studies using dermal fibroblasts from this DBP
null patient showed that DBP does not alter cellular uptake
of bioactive vitamin D and expression of the responsive gene
Frontiers in Endocrinology | www.frontiersin.org 5July 2019 | Volume 10 | Article 470
43
Kew DBP and Inflammatory Injury
CYP24A1, a very similar result also was previously reported
using cells from DBP null mice (85). However, no information
was provided about this patient’s response to infections or
tissue injury. This report provides some degree of validation
that DBP null mice can act as a model for DBP deficiency
in humans.
DBP NULL MICE AND INFLAMMATORY
INJURY MODELS
Our in vivo studies of inflammatory injury utilized the DBP
null mouse strain developed by Nancy Cooke’s lab (84,87). This
DBP null strain was re-derived the by in vitro fertilization using
DBP/sperm and a wild-type C57BL/6J female (DBP+/+)
to produce DBP+/hemizygotes which were cross-bred to
generate the DBP null (/) and wild type (+/+) mouse
colonies (81). These DBP/and DBP+/+mice have been
used in three injury models: acute lung injury, multiphasic
(acute, chronic, fibrotic) lung injury, and acute muscle injury
(81,88). Each model clearly showed that DBP null mice
always have less inflammation, and most noticeably, significantly
fewer neutrophils at the site of injury. The markedly reduced
neutrophilic inflammation observed with DBP null mice is
very consistent among the injury models and reproducible
between experiments, but the underlying cellular and molecular
mechanisms that are responsible for generating this phenotype
is only partially understood. A proposed tentative model is
described in a separate section below.
The first study to examine the in vivo role of DBP in
inflammatory injury used a model of acute complement-
dependent alveolitis, induced by either immune complexes or
purified mouse C5a (81). In both alveolitis models, DBP null
mice had significantly reduced (50%) neutrophil recruitment
to the lungs compared to their wild-type DBP+/+counterparts,
and lung histology showed significantly less inflammation in the
null mice (81). Another important observation is that addition of
exogenous DBP to the lungs of DBP null mice completely rescued
their neutrophil recruitment defect. The same study also showed
that bronchoalveolar lavage (BAL) fluid from wild-type mice had
extensive DBP-actin complexes (75% of total DBP) 4h after
induction of alveolitis. Although as predicted there were no DBP-
actin complexes in DBP null mice, there was detectable actin in
the BAL fluid from these animals, indicating actin release from
damaged cells (81). These results indicate that DBP null mice
have impaired neutrophil recruitment due to lack of DBP and not
a cellular defect since the total number, receptor expression and
chemotaxis of circulating DBP null neutrophils are essentially
identical to cells from their wild-type DBP+/+counterparts.
A second study investigated if a systemic DBP deficiency
could attenuate multiphasic lung injury and tissue remodeling
induced by bleomycin (81). Wild type and DBP null mice
received bleomycin by oropharyngeal aspiration; lung injury was
evaluated after 7, 16, or 21 days. DBP null mice all survived to
day 21 and did not display overt signs of morbidity whereas all
wild-type mice died between day 13 and 16 and showed clear
signs of respiratory distress. Bronchoalveolar lavage (BAL) fluid
from wild-type mice had extensive DBP-actin complexes (60–
75% of total DBP) whereas DBP null mice had no complexes but
had evidence of free actin. Analysis of BAL fluid on days 7 and
16 post-treatment showed that both mouse strains had similar
numbers of lung macrophages and lymphocytes, but DBP null
animals had significantly fewer lung neutrophils. Histological
analysis of the lungs on day 16 showed that DBP null mice
had a 50% decrease in fibrosis and collagen deposition as
compared wild-type animals. This study demonstrated that a
systemic deficiency in DBP provides significant protection from
bleomycin-induced inflammation and fibrosis in mice.
The third in vivo study utilized an acute muscle injury model
induced by injection of 50% glycerol into the thigh muscle (88).
All animals survived the procedure, but intramuscular glycerol
injection showed lysis of skeletal myocytes, and inflammatory
cell infiltrates in both strains of mice. The muscle inflammatory
cell infiltrate in DBP null mice had remarkably few neutrophils
as compared to wild-type mice. The neutrophil chemoattractant
CXCL1 was significantly reduced in muscle tissue from DBP null
mice. Plasma obtained 48 h after glycerol injection revealed that
DBP null mice had significantly lower levels of systemic cytokines
IL-6, CCL2, CXCL1, and G-CSF. Multiplex analysis of 36
cytokines indicated that DBP null mice had a less inflammatory
and more pro-reparative cytokine profile than their wild-type
DBP+/+counterparts (88).
These in vivo studies comparing null mice to wild-types
showed that DBP may have a central role during inflammation
since it induces selective recruitment of neutrophils, and the DBP
cofactor function is not restricted to C5a as prior in vitro studies
indicated, so the physiological implications are much broader.
So how does DBP contribute to inflammation by enhancing
neutrophil recruitment? The accumulating in vitro and in vivo
evidence appears to suggest that DBP-actin complexes may act
as an alarmin and trigger pro-inflammatory functions. DBP
is the major extracellular scavenger for actin released from
damaged/dead cells and formation of DBP-actin complexes is an
immediate host response to tissue injury. All of the in vivo injury
models discussed above had evidence of DBP-actin complexes
only in wild-type mice, and DBP repletion in the acute alveolitis
model reversed the neutrophil recruitment defect in the DBP
null mice. The currently accepted actin scavenger hypothesis
states that injury-induced depletion of plasma DBP will diminish
the actin binding capacity in blood and extracellular fluids,
leading to formation of actin filaments that obstruct flow and
damage small blood vessels (15). Furthermore, if the current actin
scavenger hypothesis is correct, it follows that DBP null mice
should succumb rapidly to an intravenous bolus of actin which
would obstruct the pulmonary vasculature. However, contrary
to what would be predicted by the actin-free DBP hypothesis,
we observed that DBP null mice do not succumb to a bolus
of purified actin injected intravenously (89). In fact, DBP null
mice were largely resistant to the lung inflammation and injury
observed in wild-type mice, and the null mice appeared utilize
plasma gelsolin to clear the actin bolus. Wild-type mice also
had a large percentage of their total plasma DBP pool (78%)
bound to actin 1.5 h after i.v. injection. Moreover, in vitro studies
showed that purified DBP-actin complexes added to cultured
Frontiers in Endocrinology | www.frontiersin.org 6July 2019 | Volume 10 | Article 470
44
Kew DBP and Inflammatory Injury
endothelial cells caused direct cell damage (at 4 h) or death (at
24 h), providing clear evidence that DBP-actin complexes have
a direct detrimental effect on cells (89). Thus, results from DBP
null mice provide strong evidence to refute the prevailing actin
scavenger hypothesis and suggest that the inverse hypothesis
may be valid, i.e., an increase in DBP-actin complexes, and
not a reduction in actin-free DBP, correlates with inflammation
and injury.
A POSSIBLE ROLE FOR VITAMIN D IN
INFLAMMATORY INJURY?
Vitamin D is known to regulate numerous genes that are involved
in the immune and inflammatory response. In vitro and in vivo
studies have shown that active vitamin D produces a tolerogenic,
anti-inflammatory, and reparative phenotype as evidenced by
immune cell activation status and cytokine profiles (90,91). DBP
null mice have almost no detectable serum vitamin D but actually
are vitamin D sufficient when fed a vitamin D replete chow
diet (84,87). Although plasma DBP does not alter the tissue
availability of vitamin D, the effect on immune cells in the blood
is not known. The lack of plasma DBP in mice may permit greater
delivery of bioactive vitamin D to immune cells during their
transit in the blood, potentially altering their transcriptomes to
dampen inflammation and limit tissue damage. Superimposed on
this possible scenario of transcriptional regulation by vitamin D
is a lack of DBP-actin complexes during tissue injury. Perhaps
both mechanisms together cause DBP null mice to have less
neutrophilic inflammation and resultant tissue damage following
injury. However, these possibilities remain to be investigated.
Finally, it is interesting to note that we previously reported that
bioactive vitamin D (1,25 dihydroxy-vitamin D3) bound to DBP
at physiologically relevant concentrations of 10 and 100 pM,
completely abolished the DBP chemotactic cofactor function of
human neutrophils in vitro, but had no effect on chemotaxis
to optimal concentrations of four different chemoattractants
(80). In contrast, 25-hydroxy-vitamin D3bound to DBP had no
effect on the chemotactic cofactor function (80), thus providing
evidence of a direct inhibitory effect of bioactive vitamin D on the
DBP chemotactic cofactor function for neutrophils.
PROPOSED MODEL OF DBP-ACTIN
COMPLEXES AND INFLAMMATORY
INJURY
Recent evidence indicates that DBP bound to G-actin, and not
free DBP, functions as an indirect but essential cofactor for
neutrophil migration, thus, providing a possible mechanism to
explain how DBP functions to enhance neutrophil migration in
vitro and recruitment to sites of inflammation in vivo (81,88,
89). However, it is not clear how DBP-actin complexes enhance
neutrophil recruitment and inflammation. It is interesting to
speculate that DBP-actin complexes may augment a chemotactic
signal and cause release of other proinflammatory molecules
stored within neutrophils. Perhaps the most attractive candidate
in this scenario is calprotectin, a 24 kDa heterodimer composed
of S100A8 and S100A9 that can be rapidly released from
neutrophils (92). Abundant quantities of S100A8 and S100A9
are stored in the cytosol of neutrophils, and upon cell activation
these molecules can be released into the extracellular space
as active heterodimers or heterotetramers (92). S100A8/A9
has multiple proinflammatory functions and has been shown
to mediate both neutrophil bone marrow development and
facilitate chemotaxis of mature circulating cells, most likely by
binding to toll-like receptor 4 (TLR4) on the plasma membrane
(92). The proposed mechanism involving DBP-actin complexes
has clear physiological relevance since DBP is abundant and
ubiquitous in all fluid compartments, and release of G-actin
from damaged/dead cells is a consistent feature in all types
of inflammatory injury. Moreover, we have previously reported
that DBP binds to actin on the neutrophil plasma membrane
followed by elastase-mediated shedding of these complexes,
perhaps as microvesicles, into the extracellular fluids (46,48,49).
We propose that DBP-actin complexes can bind (or re-bind
following shedding) to a neutrophil surface receptor that triggers
S100A8/A9 release. In turn, S100A8/A9 binds in an autocrine
or paracrine manner to neutrophil TLR4 inducing a signal that
synergizes with the chemoattractant receptor signal to enhance
migration. Furthermore, it is well-known that TLR4 ligation and
signaling synergizes with signals from chemoattractant receptors
to enhance leukocyte chemotaxis both in vitro and in vivo
(93,94).
This provisional model may explain how a deficiency of
DBP results in significantly decreased neutrophilic inflammation.
However, there are several “unknowns” with this model that need
to be investigated. First, the putative receptor for binding DBP-
actin complexes has not been identified, but other studies have
shown that the receptor Clec9A binds F-actin and is involved
in sensing damaged cells (9597). It is not known if the Clec9A
receptor also binds G-actin complexed with DBP or if even
if this receptor is expressed on neutrophils. Second, it is not
known if the putative DBP-actin receptor signals release of
S100A8/A9 when it is ligated with complexes. Third, a question
remains do all DBP-actin complexes function the same or are
there modifications that differentiate between inflammatory and
benign complexes. Finally, do other cell types besides neutrophils
and endothelial cells also respond to DBP-actin complexes,
particularly hepatocytes and Kupffer cells in the liver (the
primary site of DBP-actin clearance).
FUTURE DIRECTIONS
Many questions about the functions of DBP remain to be
answered. However, new investigative tools and experimental
approaches will be needed to decipher these functions. For
example, new mouse models with tissue specific inducible
expression of DBP, or a mouse model constructed with
selective deletions in functional regions within DBP (vitamin
D, actin, cell binding regions) by utilizing CRISPER-Cas9
gene editing technology. Another essential tool currently
needed is an antibody that only recognizes a neoepitope on
DBP-actin complexes, and not DBP or G-actin monomers.
Frontiers in Endocrinology | www.frontiersin.org 7July 2019 | Volume 10 | Article 470
45
Kew DBP and Inflammatory Injury
This antibody then could be used to develop an ELISA
that specifically detects only DBP-actin complexes in
biological fluids. In closing, research into the biological
functions of DBP has progressed far since Hirschfeld’s initial
description 60 years ago, but perhaps in the near future
new experimental approaches using advanced technologies
(single cell transcriptomics, mass cytometry, advanced
microscopy, and in vivo imaging, etc.) and bioinformatic
analysis may reveal what DBP actually has been doing
all along.
AUTHOR CONTRIBUTIONS
The author confirms being the sole contributor of this work and
has approved it for publication.
ACKNOWLEDGMENTS
We would like to acknowledge the long-term grant support
from the U.S. National Institutes of Health (R01GM063769) that
facilitated these investigations.
REFERENCES
1. Hirschfeld J. Immunoelectrophoretic demonstration of qualatative differences
in normal human sera and their relation to the haptoglobins. Acta Path
Microbiol Scand. (1959) 47:160. doi: 10.1111/j.1699-0463.1959.tb04844.x
2. Hirschfeld J, Jonsson B, Rasmuson M. Inheritance of a new group-specific
system demonstrated in normal human sera by means of an immuno-
electrophoretic technique. Nature. (1960) 185:931–2. doi: 10.1038/185931b0
3. Hirschfeld J. Immuno-electrophoretic differentiation of haptoglobins from
another group-specific inheritable system in normal human sera. Nature.
(1960) 187:126–8. doi: 10.1038/187126a0
4. Daiger SP, Schanfield MS, Cavalli-Sforza LL. Group-specific component (Gc)
proteins bind vitamin D and 25-hydroxyvitamin D. Proc Natl Acad Sci USA.
(1975) 72:2076–80. doi: 10.1073/pnas.72.6.2076
5. Cooke NE, Walgate J, Haddad JG Jr. Human serum binding protein
for vitamin D and its metabolites. I. Physicochemical and immunological
identification in human tissues. J Biol Chem. (1979) 254:5958–64.
6. Cooke NE, Walgate J, Haddad JG Jr. Human serum binding protein for
vitamin D and its metabolites. II. Specific, high affinity association with a
protein in nucleated tissue. J Biol Chem. (1979) 254:5965–71.
7. Van Baelen H, Bouillon R, De Moor P. Binding of 25-hydroxycholecalciferol
in tissues. J Biol Chem. (1977) 252:2515–8.
8. Van Baelen H, Bouillon R, De Moor P. Vitamin D-binding protein (Gc-
globulin) binds actin. J Biol Chem. (1980) 255:2270–2.
9. Verboven C, Rabijns A, De Maeyer M, Van Baelen H, Bouillon R, De Ranter
C. A structural basis for the unique binding features of the human vitamin
D-binding protein. Nat Struct Biol. (2002) 9:131–6. doi: 10.1038/nsb754
10. Verboven C, Bogaerts I, Waelkens E, Rabijns A, Van Baelen H, Bouillon R,
et al. Actin-DBP: the perfect structural fit? Acta Crystallogr D Biol Crystallogr.
(2003) 59:263–73. doi: 10.1107/S0907444902021455
11. Delanghe JR, Speeckaert R, Speeckaert MM. Behind the scenes of vitamin D
binding protein: more than vitamin D binding. Best Pract Res Clin Endocrinol
Metabol. (2015) 29:773–86. doi: 10.1016/j.beem.2015.06.006
12. Goldschmidt-Clermont PJ, Lee WM, Galbraith RM. Proportion of circulating
Gc (vitamin D-binding protein) in complexed form: relation to clinical
outcome in fulminant hepatic necrosis. Gastroenterology. (1988) 94:1454–8.
doi: 10.1016/0016-5085(88)90686-5
13. Dahl B, Schiodt FV, Ott P, Wians F, Lee WM, Balko J, et al.
Plasma concentration of Gc-globulin is associated with organ
dysfunction and sepsis after injury. Crit Care Med. (2003) 31:152–6.
doi: 10.1097/00003246-200301000-00024
14. Antoniades CG, Berry PA, Bruce M, Cross TJ, Portal AJ, Hussain MJ, et al.
Actin-free Gc globulin: a rapidly assessed biomarker of organ dysfunction
in acute liver failure and cirrhosis. Liver Transplant. (2007) 13:1254–61.
doi: 10.1002/lt.21196
15. Meier U, Gressner O, Lammert F, Gressner AM. Gc-globulin: roles in response
to injury. Clin Chem. (2006) 52:1247–53. doi: 10.1373/clinchem.2005.065680
16. Shen H, Kreisel D, Goldstein DR. Processes of sterile inflammation. J
Immunol. (2013) 191:2857–63. doi: 10.4049/jimmunol.1301539
17. dos Remedios CG, Chhabra D, Kekic M, Dedova IV, Tsubakihara M, Berry
DA, et al. Actin binding proteins: regulation of cytoskeletal microfilaments.
Physiol Rev. (2003) 83:433–73. doi: 10.1152/physrev.00026.2002
18. Rottner K, Stradal TE. Actin dynamics and turnover in cell motility. Curr Opin
Cell Biol. (2011) 23:569–78. doi: 10.1016/j.ceb.2011.07.003
19. Haddad JG, Harper KD, Guoth M, Pietra GG, Sanger JW. Angiopathic
consequences of saturating the plasma scavenger system for actin. Proc Natl
Acad Sci USA. (1990) 87:1381–5. doi: 10.1073/pnas.87.4.1381
20. Chun RF. New perspectives on the vitamin D binding protein. Cell Biochem
Funct. (2012) 30:445–56. doi: 10.1002/cbf.2835
21. Malik S, Fu L, Juras DJ, Karmali M, Wong BY, Gozdzik A, et al. Common
variants of the vitamin D binding protein gene and adverse health outcomes.
Crit Rev Clin Lab Sci. (2013) 50:1–22. doi: 10.3109/10408363.2012.750262
22. Swamy N, Head JF, Weitz D, Ray R. Biochemical and preliminary
crystallographic characterization of the vitamin D sterol- and actin-binding by
human vitamin D-binding protein. Arch Biochem Biophys. (2002) 402:14–23.
doi: 10.1016/S0003-9861(02)00033-4
23. Otterbein LR, Cosio C, Graceffa P, Dominguez R. Crystal structures of
the vitamin D-binding protein and its complex with actin: Structural basis
of the actin-scavenger system. Proc Natl Acad Sci USA. (2002) 99:8003–8.
doi: 10.1073/pnas.122126299
24. Haddad JG, Hu YZ, Kowalski MA, Laramore C, Ray K, Robzyk P,
et al. Identification of the sterol- and actin-binding domains of plasma
vitamin D binding protein (Gc-globulin). Biochemistry. (1992) 31:7174–81.
doi: 10.1021/bi00146a021
25. Cooke NE, Haddad JG. Vitamin D binding protein (Gc-globulin). Endocr Rev.
(1989) 10:294–307. doi: 10.1210/edrv-10-3-294
26. White P, Cooke N. The multifunctional properties and characteristics of
vitamin D-binding protein. Trends Endocrinol Metabol. (2000) 11:320–7.
doi: 10.1016/S1043-2760(00)00317-9
27. Bouillon R, Van Assche FA, Van Baelen H, Heyns W, De Moor P. Influence
of the vitamin D-binding protein on the serum concentration of 1,25-
dihydroxyvitamin D3. Significance of the free 1,25-dihydroxyvitamin D3
concentration. J Clin Invest. (1981) 67:589–96. doi: 10.1172/JCI110072
28. Harper KD, McLeod JF, Kowalski MA, Haddad JG. Vitamin D binding protein
sequesters monomeric actin in the circulation of the rat. J Clin Invest. (1987)
79:1365. doi: 10.1172/JCI112963
29. Xiao W, Mindrinos MN, Seok J, Cuschieri J, Cuenca AG, Gao H, et al. A
genomic storm in critically injured humans. J Exp Med. (2011) 208:2581–90.
doi: 10.1084/jem.20111354
30. Petrini M, Emerson DL, Galbraith RM. Linkage between surface
immunoglobulin and cytoskeleton of B lymphocytes may involve Gc
protein. Nature. (1983) 306:73–4. doi: 10.1038/306073a0
31. Petrini M, Galbraith RM, Werner PAM, Emerson DL, Arnaud
P. Gc (Vitamin D Binding Protein) binds to cytoplasm of all
human lymphocytes and is expressed on B-cell membranes. Clin
Immunol Immunopathol. (1984) 31:282–95. doi: 10.1016/0090-1229(84)
90248-4
32. Petrini M, Allegrini A, Ambrogi F, Valentini P, Sabbatini A, Arnaud P,
et al. Binding of Gc (VDBP) to membranes of human B lymphocytes
following stripping of extant protein. J Endocrinol Invest. (1995) 18:630–7.
doi: 10.1007/BF03349781
33. Petrini M, Galbraith RM, Emerson DL, Nel AE, Arnaud P. Structural studies
of T lymphocyte Fc receptors. Association of Gc protein with IgG binding to
Fc gamma. J Biol Chem. (1985) 260:1804–10.
Frontiers in Endocrinology | www.frontiersin.org 8July 2019 | Volume 10 | Article 470
46
Kew DBP and Inflammatory Injury
34. Yu HM, Li XJ, Kadam AL, Cheng CY, Koide SS. Human testis vitamin
D binding protein involved in infertility. Arch Androl. (1994) 33:119–28.
doi: 10.3109/01485019408987813
35. Nestler JE, McLeod JF, Kowalski MA, Strauss JF, Haddad JG. Detection
of Vitamin D binding protein on the surface of cytotrophoblasts
isolated from human placentae. Endocrinology. (1987) 120:1996–2002.
doi: 10.1210/endo-120-5-1996
36. Emerson DL, Werner PA, Cheng MH, Galbraith RM. Presence
of Gc (Vitamin D-Binding Protein) and interactions with
actin in human placental tissue. Am J ReprodImmunol. (1985)
7:15–21. doi: 10.1111/j.1600-0897.1985.tb00257.x
37. Wooten MW, Nel AE, Goldschmidt-Clermont PJ, Galbraith RM, Wrenn
RW. Identification of a major endogenous substrate for phospholipid/Ca2+-
dependent kinase in pancreatic acini as Gc (vitamin D-binding protein). FEBS
Lett. (1985) 191:97–101. doi: 10.1016/0014-5793(85)81001-2
38. McLeod JF, Kowalski MA, Haddad JG. Characterization of a monoclonal
antibody to human serum vitamin D binding protein (Gc globulin):
recognition of an epitope hidden in membranes of circulating monocytes.
Endocrinology. (1986) 119:77–83. doi: 10.1210/endo-119-1-77
39. Kew RR, Sibug MA, Liuzzo JP, Webster RO. Localization and quantitation of
the vitamin D binding protein (Gc-globulin) in human neutrophils. Blood.
(1993) 82:274–83.
40. Keenan MJ, Holmes RP. The uptake and metabolism of
25-hydroxyvitamin D3 and vitamin D binding protein by
cultured porcine kidney cells (LLC-PK1). Int J Biochem. (1991)
23:1225–30. doi: 10.1016/0020-711X(91)90220-H
41. Guoth M, Murgia A, Smith RM, Prystowsky MB, Cooke NE, Haddad JG. Cell
surface vitamin D-binding protein (Gc-globulin) is acquired from plasma.
Endocrinology. (1990) 127:2313–21. doi: 10.1210/endo-127-5-2313
42. Nykjaer A, Dragun D, Walther D, Vorum H, Jacobsen C, Herz
J, et al. An endocytic pathway essential for renal uptake and
activation of the steroid 25-(OH) vitamin D3. Cell. (1999) 96:507–15.
doi: 10.1016/S0092-8674(00)80655-8
43. Nykjaer A, Fyfe JC, Kozyraki R, Leheste JR, Jacobsen C, Nielsen MS, et al.
Cubilin dysfunction causes abnormal metabolism of the steroid hormone
25(OH) vitamin D(3). Proc Natl Acad Sci USA. (2001) 98:13895–900.
doi: 10.1073/pnas.241516998
44. Leheste JR, Rolinski B, Vorum H, Hilpert J, Nykjaer A, Jacobsen
C, et al. Megalin knockout mice as an animal model of low
molecular weight proteinuria. Am J Pathol. (1999) 155:1361–70.
doi: 10.1016/S0002-9440(10)65238-8
45. Saito A, Iino N, Takeda T, Gejyo F. Role of megalin, a proximal tubular
endocytic receptor, in calcium and phosphate homeostasis. Therap Apheresis
Dial. (2007) 11(Suppl. 1):S23–6. doi: 10.1111/j.1744-9987.2007.00514.x
46. DiMartino SJ, Kew RR. Initial characterization of the vitamin D
binding protein (Gc-globulin) binding site on the neutrophil plasma
membrane: evidence for a chondroitin sulfate proteoglycan. J Immunol.
(1999) 163:2135–42.
47. McVoy LA, Kew RR. CD44 and annexin A2 mediate the C5a chemotactic
cofactor function of the vitamin D binding protein. J Immunol. (2005)
175:4754–60. doi: 10.4049/jimmunol.175.7.4754
48. DiMartino SJ, Trujillo G, McVoy LA, Zhang J, Kew RR. Upregulation of
vitamin D binding protein (Gc-globulin) binding sites during neutrophil
activation from a latent reservoir in azurophil granules. Mol Immunol. (2007)
44:2370–7. doi: 10.1016/j.molimm.2006.10.011
49. DiMartino SJ, Shah AB, Trujillo G, Kew RR. Elastase controls the binding of
the vitamin D-binding protein (Gc-globulin) to neutrophils: a potential role in
the regulation of C5a co-chemotactic activity. J Immunol. (2001) 166:2688–94.
doi: 10.4049/jimmunol.166.4.2688
50. Zhang J, Habiel DM, Ramadass M, Kew RR. Identification of two distinct cell
binding sequences in the vitamin D binding protein. Biochim Biophys Acta.
(2010) 1803:623–9. doi: 10.1016/j.bbamcr.2010.02.010
51. Liu Y, Myrvang HK, Dekker LV. Annexin A2 complexes with S100 proteins:
structure, function and pharmacological manipulation. Br J Pharmacol. (2015)
172:1664–76. doi: 10.1111/bph.12978
52. Amulic B, Cazalet C, Hayes GL, Metzler KD, Zychlinsky A. Neutrophil
function: from mechanisms to disease. Annu Rev Immunol. (2012) 30:459–89.
doi: 10.1146/annurev-immunol-020711-074942
53. Campbell EJ, Campbell MA, Boukedes SS, Owen CA. Quantum proteolysis by
neutrophils: implications for pulmonary emphysema in alpha 1-antitrypsin
deficiency. J Clin Invest. (1999) 104:337–44. doi: 10.1172/JCI6092
54. Owen CA, Campbell MA, Sannes PL, Boukedes SS, Campbell EJ.
Cell surface-bound elastase and cathepsin G on human neutrophils: A
novel, non-oxidative mechanism by which neutrophils focus and preserve
catalytic activity of serine proteinases. J Cell Biol. (1995) 131:775–89.
doi: 10.1083/jcb.131.3.775
55. Borregaard N, Cowland JB. Granules of the human neutrophilic
polymorphonuclear leukocyte. Blood. (1997) 89:3503–21.
56. Lawrence SM, Corriden R, Nizet V. The Ontogeny of a Neutrophil:
Mechanisms of Granulopoiesis and Homeostasis. Microbiol Mol Biol Rev.
(2018) 82:e00057–17. doi: 10.1128/MMBR.00057-17
57. Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and
inflammation. Nat Rev Immunol. (2013) 13:159–75. doi: 10.1038/nri3399
58. Mocsai A. Diverse novel functions of neutrophils in immunity, inflammation,
and beyond. J Exp Med. (2013) 210:1283–99. doi: 10.1084/jem.20122220
59. Borregaard N. Neutrophils, from marrow to microbes. Immunity. (2010)
33:657–70. doi: 10.1016/j.immuni.2010.11.011
60. Wirths S, Bugl S, Kopp HG. Neutrophil homeostasis and
its regulation by danger signaling. Blood. (2014) 123:3563–6.
doi: 10.1182/blood-2013-11-516260
61. Manz MG, Boettcher S. Emergency granulopoiesis. Nat Rev Immunol. (2014)
14:302–14. doi: 10.1038/nri3660
62. Nathan C. Neutrophils and immunity: challenges and opportunities. Nat Rev
Immunol. (2006) 6:173–82. doi: 10.1038/nri1785
63. Ley K, Hoffman HM, Kubes P, C assatella MA, ZychlinskyA, Hedrick C C, et al.
Neutrophils: New insights and open questions. Science immunology. (2018)
3:eaat4579. doi: 10.1126/sciimmunol.aat4579
64. Schultze JL, Mass E, Schlitzer A. Emerging principles in myelopoiesis
at homeostasis and during infection and inflammation. Immunity. (2019)
50:288–301. doi: 10.1016/j.immuni.2019.01.019
65. Ng LG, Ostuni R, Hidalgo A. Heterogeneity of neutrophils. Nat Rev Immunol.
(2019) 19:255–65. doi: 10.1038/s41577-019-0141-8
66. Liew PX, Kubes P. The neutrophil’s role during health and disease. Physiol Rev.
(2019) 99:1223–48. doi: 10.1152/physrev.00012.2018
67. Fernandez HN, Henson PM, Otani A, Hugli TE. Chemotactic response
to human C3a and C5a anaphylatoxins. I. Evaluation of C3a and C5a
leukotaxis in vitro and under simulated in vivo conditions. J Immunol.
(1978) 120:109–15.
68. Beebe DP, Ward PA, Spitznagel JK. Isolation and characterization of an acidic
chemotactic factor from complement-activated human serum. Clin Immunol
Immunopathol. (1980) 15:88–105. doi: 10.1016/0090-1229(80)90023-9
69. Perez HD, Goldstein IM, Chernoff D, Webster RO, Henson PM. Chemotactic
activity of C5a des Arg: evidence of a requirement for an anioinic
peptide “helper factor” and inhibition by a cationic protein in serum from
patients with systemic lupus erythematosus. Mol Immunol. (1980) 17:163–9.
doi: 10.1016/0161-5890(80)90068-1
70. Perez HD, Goldstein IM, Webster RO, Henson PM. Enhancement of the
chemotactic activity of human C5a des Arg by an anionic polypeptide
(“cochemotaxin”) in normal serum and plasma. J Immunol. (1981) 126:800–4.
71. Kew RR, Webster RO. Gc-globulin (vitamin D-binding protein) enhances the
neutrophil chemotactic activity of C5a and C5a des Arg. J Clin Invest. (1988)
82:364–9. doi: 10.1172/JCI113596
72. Perez HD, Kelly E, Chenoweth D, Elfman F. Identification of the C5a des Arg
cochemotaxin. Homology with vitamin D-binding protein (group-specific
component globulin). J Clin Invest. (1988) 82:360–3. doi: 10.1172/JCI113595
73. Senior RM, Griffin GL, Perez HD, Webster RO. Human C5a and C5a des Arg
exhibit chemotactic activity for fibroblasts. J Immunol. (1988) 141:3570–4.
74. Robbins RA, Hamel FG. Chemotactic factor inactivator interaction with
Gc-globulin (vitamin D-binding protein). A mechanism of modulating the
chemotactic activity of C5a. J Immunol. (1990) 144:2371–6.
75. Petrini M, Azzara A, Carulli G, Ambrogi F, Galbraith RM. 1,25-
dihydroxycholecalciferol inhibits the cochemotactic activity of Gc
(vitamin D binding protein). J Endocrinol Invest. (1991) 14:405–8.
doi: 10.1007/BF03349090
76. Metcalf JP, Thompson AB, Gossman GL, Nelson KJ, Koyama S, Rennard SI,
et al. Gc-globulin functions as a cochemotaxin in the lower respiratory tract.
Frontiers in Endocrinology | www.frontiersin.org 9July 2019 | Volume 10 | Article 470
47
Kew DBP and Inflammatory Injury
A potential mechanism for lung neutrophil recruitment in cigarette smokers.
Am Rev Respir Dis. (1991) 143:844–9. doi: 10.1164/ajrccm/143.4_Pt_1.844
77. Zwahlen RD, Roth DR. Chemotactic competence of neutrophils from
neonatal calves. Functional comparison with neutrophils from adult cattle.
Inflammation. (1990) 14:109–23. doi: 10.1007/BF00914034
78. Piquette CA, Robinson-Hill R, Webster RO. Human monocyte chemotaxis
to complement-derived chemotaxins is enhanced by Gc-globulin. J Leukocyte
Biol. (1994) 55:349–54. doi: 10.1002/jlb.55.3.349
79. Binder R, Kress A, Kan G, Herrmann K, Kirschfink M. Neutrophil priming
by cytokines and vitamin D binding protein (Gc-globulin): impact on C5a-
mediated chemotaxis, degranulation and respiratory burst. Mol Immunol.
(1999) 36:885–92. doi: 10.1016/S0161-5890(99)00110-8
80. Shah AB, DiMartino SJ, Trujillo G, Kew RR. Selective inhibition
of the C5a chemotactic cofactor function of the vitamin D binding
protein by 1,25(OH)2 vitamin D3. Mol Immunol. (2006) 43:1109–15.
doi: 10.1016/j.molimm.2005.07.023
81. Trujillo G, Habiel DM, Ge L, Ramadass M, Cooke NE, Kew RR. Neutrophil
recruitment to the lung in both C5a- and CXCL1-induced alveolitis is
impaired in vitamin D-binding protein-deficient mice. J Immunol. (2013)
191:848–56. doi: 10.4049/jimmunol.1202941
82. Kew RR, Mollison KW, Webster RO. Binding of Gc globulin (vitamin D
binding protein) to C5a or C5a des Arg is not necessary for co-chemotactic
activity. J Leukoc Biol. (1995) 58:55–8. doi: 10.1002/jlb.58.1.55
83. Perez HD. Gc globulin (vitamin D-binding protein) increases binding of low
concentrations of C5a des Arg to human polymorphonuclear leukocytes: An
explanation for its cochemotaxin activity. Inflammation. (1994) 18:215–20.
doi: 10.1007/BF01534562
84. Safadi FF, Thornton P, Magiera H, Hollis BW, Gentile M, Haddad JG, et al.
Osteopathy and resistance to vitamin D toxicity in mice null for vitamin D
binding protein. J Clin Invest. (1999) 103:239–51. doi: 10.1172/JCI5244
85. 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) 149:3656–67. doi: 10.1210/en.2008-0042
86. Henderson CM, Fink SL, Bassyouni H, Argiropoulos B, Brown L, Laha TJ,
et al. Vitamin D-Binding Protein Deficiency and Homozygous Deletion of the
GC Gene. N Engl J Med. (2019) 380:1150–7. doi: 10.1056/NEJMoa1807841
87. White P, Liebhaber SA, Cooke NE. 129X1/SvJ mouse strain has a novel
defect in inflammatory cell recruitment. J Immunol. (2002) 168:869–74.
doi: 10.4049/jimmunol.168.2.869
88. Kew RR, Tabrizian T, Vosswinkel JA, Davis JE, Jawa RS. Vitamin D-binding
protein deficiency in mice decreases systemic and select tissue levels of
inflammatory cytokines in a murine model of acute muscle injury. J Trauma
Acute Care Surg. (2018) 84:847–54. doi: 10.1097/TA.0000000000001875
89. Ge L, Trujillo G, Miller EJ, Kew RR. Circulating complexes of the
vitamin D binding protein with G-actin induce lung inflammation
by targeting endothelial cells. Immunobiology. (2014) 219:198–207.
doi: 10.1016/j.imbio.2013.10.001
90. Wang TT, Tavera-Mendoza LE, Laperriere D, Libby E, MacLeod NB, Nagai
Y, et al. Large-scale in silico and microarray-based identification of direct
1,25-dihydroxyvitamin D3 target genes. Mol Endocrinol. (2005) 19:2685–95.
doi: 10.1210/me.2005-0106
91. White JH. Vitamin D metabolism and signaling in the immune system. Rev
Endocr Metab Disord. (2012) 13:21–9. doi: 10.1007/s11154-011-9195-z
92. Wang S, Song R, Wang Z, Jing Z, Wang S, Ma J. S100A8/A9 in Inflammation.
Front Immunol. (2018) 9:1298. doi: 10.3389/fimmu.2018.01298
93. Liu Z, Jiang Y, Li Y, Wang J, Fan L, Scott MJ, et al. TLR4 Signaling augments
monocyte chemotaxis by regulating G protein-coupled receptor kinase 2
translocation. J Immunol. (2013) 191:857–64. doi: 10.4049/jimmunol.13
00790
94. Aomatsu K, Kato T, Fujita H, Hato F, Oshitani N, Kamata N, et al. Toll-
like receptor agonists stimulate human neutrophil migration via activation
of mitogen-activated protein kinases. Immunology. (2008) 123:171–80.
doi: 10.1111/j.1365-2567.2007.02684.x
95. Del Fresno C, Saz-Leal P, Enamorado M, Wculek SK,
Martinez-Cano S, Blanco-Menendez N, et al. DNGR-1 in
dendritic cells limits tissue damage by dampening neutrophil
recruitment. Science. (2018) 362:351–6. doi: 10.1126/science.
aan8423
96. Ahrens S, Zelenay S, Sancho D, Hanc P, Kjaer S, Feest C, et al. F-
actin is an evolutionarily conserved damage-associated molecular pattern
recognized by DNGR-1, a receptor for dead cells. Immunity. (2012) 36:635–45.
doi: 10.1016/j.immuni.2012.03.008
97. Zhang JG, Czabotar PE, Policheni AN, Caminschi I, Wan SS, Kitsoulis S,
et al. The dendritic cell receptor Clec9A binds damaged cells via exposed
actin filaments. Immunity. (2012) 36:646–57. doi: 10.1016/j.immuni.2012.
03.009
Conflict of Interest Statement: The author declares that the research was
conducted in the absence of any commercial or financial relationships that could
be construed as a potential conflict of interest.
Copyright © 2019 Kew. This is an open-access article distributed under the terms
of the Creative Commons Attribution License (CC BY). The use, distribution or
reproduction in other forums is permitted, provided the original author(s) and the
copyright owner(s) are credited and that the original publication in this journal
is cited, in accordance with accepted academic practice. No use, distribution or
reproduction is permitted which does not comply with these terms.
Frontiers in Endocrinology | www.frontiersin.org 10 July 2019 | Volume 10 | Article 470
48
REVIEW
published: 24 October 2019
doi: 10.3389/fendo.2019.00718
Frontiers in Endocrinology | www.frontiersin.org 1October 2019 | Volume 10 | Article 718
Edited by:
Zhongjian Xie,
Central South University, China
Reviewed by:
Michael F. Holick,
Boston Medical Center, United States
Melissa Orlandin Premaor,
Federal University of Minas
Gerais, Brazil
*Correspondence:
Rene F. Chun
rchun@mednet.ucla.edu
Specialty section:
This article was submitted to
Bone Research,
a section of the journal
Frontiers in Endocrinology
Received: 26 April 2019
Accepted: 04 October 2019
Published: 24 October 2019
Citation:
Chun RF, Shieh A, Gottlieb C,
Yacoubian V, Wang J, Hewison M and
Adams JS (2019) Vitamin D Binding
Protein and the Biological Activity of
Vitamin D. Front. Endocrinol. 10:718.
doi: 10.3389/fendo.2019.00718
Vitamin D Binding Protein and the
Biological Activity of Vitamin D
Rene F. Chun 1
*, Albert Shieh 2, Carter Gottlieb 1, Vahe Yacoubian 1, Jeffrey Wang 1,
Martin Hewison 3and John S. Adams 1
1Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States,
2Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States, 3Institute of
Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
Vitamin D has a long-established role in bone health. In the last two decades, there
has been a dramatic resurgence in research interest in vitamin D due to studies that
have shown its possible benefits for non-skeletal health. Underpinning the renewed
interest in vitamin D was the identification of the vital role of intracrine or localized,
tissue-specific, conversion of inactive pro-hormone 25-hydroxyvitamin D [25(OH)D] to
active 1,25-dihydroxyvitamin D [1,25(OH)2D]. This intracrine mechanism is the likely
driving force behind vitamin D action resulting in positive effects on human health.
To fully capture the effect of this localized, tissue-specific conversion to 1,25(OH)2D,
adequate 25(OH)D would be required. As such, low serum concentrations of 25(OH)D
would compromise intracrine generation of 1,25(OH)2D within target tissues. Consistent
with this is the observation that all adverse human health consequences of vitamin D
deficiency are associated with a low serum 25(OH)D level and not with low 1,25(OH)2D
concentrations. Thus, clinical investigators have sought to define what concentration
of serum 25(OH)D constitutes adequate vitamin D status. However, since 25(OH)D is
transported in serum bound primarily to vitamin D binding protein (DBP) and secondarily
to albumin, is the total 25(OH)D (bound plus free) or the unbound free 25(OH)D the crucial
determinant of the non-classical actions of vitamin D? While DBP-bound-25(OH)D is
important for renal handling of 25(OH)D and endocrine synthesis of 1,25(OH)2D, how
does DBP impact extra-renal synthesis of 1,25(OH)2D and subsequent 1,25(OH)2D
actions? Are their pathophysiological contexts where total 25(OH)D and free 25(OH)D
would diverge in value as a marker of vitamin D status? This review aims to introduce and
discuss the concept of free 25(OH)D, the molecular biology and biochemistry of vitamin
D and DBP that provides the context for free 25(OH)D, and surveys in vitro, animal, and
human studies taking free 25(OH)D into consideration.
Keywords: vitamin D, free vitamin D, bone, immunology, DBP, CYP27B1, VDR
INTRODUCTION
The benefits of vitamin D for mineral homeostasis and bone health are well-established. Deficiency
of vitamin D, rickets in children and osteomalacia in adults, can be treated or prevented with oral
supplements of vitamin D. Despite promising pre-clinical observations and vitamin D-deficiency
association studies, the impact of vitamin D on other aspects of human health such as common
49
Chun et al. Free 25D
cancers, cardiovascular disease, type 2 diabetes obesity,
autoimmune disorders, and infectious disease remains
controversial (15). Randomized, controlled supplementation
trials are required to better define the extra-skeletal roles
of vitamin D. However, these trials are complicated by two
unanswered questions: (1) what is the best marker of vitamin
D status and (2) what constitutes a level that is sufficient to
promote the health benefits of vitamin D?
ENDOCRINE VITAMIN D METABOLISM
AND ACTION
The name “vitamin D” in this review refers to a collection
of secosterol molecules detectable in the serum of vertebrates
(left panel, Figure 1). Briefly, cholecalciferol or vitamin D3
(vitamin D) results from the ultraviolet B (UVB; 290–315 nm)-
mediated photolytic-conversion of 7-dehydrocholesterol (DHC)
in skin (68). Vitamin D can also be obtained from (i)
food, principally from fortified dairy and juice products, (ii)
consumption of fresh caught fish (e.g., salmon) (9), and (iii) oral
vitamin D supplements. Vitamin D2 (ergocalciferol) is naturally
found in fungi (e.g., mushrooms) and sometimes used in food
fortification and supplementation regimes. Regardless vitamin
D2 proceeds through the same modifications as described for
vitamin D3 below.
Once in the general circulation vitamin D is bound to its
serum carrier, vitamin D binding protein (DBP) and to a lesser
extent albumin (10,11), and is subject to a first hydroxylation
step by vitamin D substrate-dependent 25-hydroxylase [CYP2R1
(12) and possibly a yet unidentified hydroxylase(s) (13)] in
the liver resulting in 25-hydroxyvitamin D (25(OH)D). Very
little 25(OH)D (5%) is secreted in to the bile (14). Rather,
the bulk of 25(OH)D re-enters the circulation, once again
bound to either DBP or albumin for endocrine transport to
target tissues. DBP- and albumin-bound 25(OH)D in urine
is reclaimed by tubular epithelial cells in the kidney (15).
Here internalized 25(OH)D is freed from its carrier protein(s),
becoming substrate for (i) the low capacity 25-hydroxyvitamin
D-1-alpha-hydroxylase (CYP27B1) and production of the
active, hormonal form of vitamin D, 1,25-dihydroxyvitamin D
(1,25(OH)2D) or (ii) the high calpacity 25-dihydroxyvitamin D-
24 hyxdoxylase (CYP24A1) to form the largely non-biologically
active metabolites 24,25-dihydroxyvitamin D (24,25(OH)2D)
and 1,24,25-trihydroxyvitamin D, respectively (16). The various
hydoxylated forms gain access to the general circulation
bound to DBP or albumin. Rheostatic endocrine control
over the reciprocal production of 1- and 24-hydroxylated
vitamin D metabolites is exerted by parathyroid hormone and
FGF23. Parathyroid hormone increases activity of CYP27B1-
hydroxylase (17,18), decreases product output by CYP24A1
(19,20); thus, increases the “activation” quotient of product
1,25(OH)2D:substrate 25(OH)D in the serum. On the other
hand, FGF-23 blunts CYP27B1 activity (21) and promotes
CYP24A1 activity; thus, decreases the 1,25(OH)2D:25(OH)D
activation quotient and increases the 24,25(OH)2D:25(OH)D
inactivation quotient (22,23).
As noted above, 1,25(OH)2D can be chaperoned in an
endocrine mode to potential target tissues that employ serum
bound, extracellular 1,25(OH)2D as a specific ligand for
transactivation of the vitamin D receptor (VDR) in the target cell
driving 1,25(OH)2D-VDR directed differential gene expression
(24). A major caveat in the concept of direct endocrine action
of 1,25(OH)2Dis the fact all adverse physiological consequences
of vitamin D deficiency in humans are associated with a low
serum 25(OH)D, not a low 1,25(OH)2D level [Table 1; (25)].
In fact, in the basal state, before treatment to raise 25D levels,
subjects with low serum 25D often have serum 1,25D levels
that are relatively elevated as a consequence of compensatory
secondary hyperparathyroidism. In this instance the increase in
the host’s circulating concentration of PTH drives an increase in
renal 1,25D production. This suggests that 25D deficiency in the
serum is a cause for “endocrine resistance” to circulating levels
of the 1,25D hormone at the level of the gut. After vitamin D
restoration treatment with return of 25D balance to normal and
resolution of secondary hyperparathyroidism, there is an increase
in intestinal calcium absorption even though there is a relative
decrease, or no change in the circulating serum 1,25D level. This
suggests that there may be local conversion of 25D to 1,25D in the
gut outside of the serum compartment that is driving intestinal
calcium absorption and/or that only measuring the total amount
of vitamin D metabolite(s) in the serum, may be an inadequate
biomarker of response to restoration of 25D levels in the blood
to normal.
LOCAL VITAMIN D METABOLISM AND
ACTION
In non-renal tissues, the CYP27B1 converts 25(OH)D to
1,25(OH)2D for local usage in paracrine, autocrine, and
intracrine regulated activities (26). Perhaps the most relevant
physiological/pathophysiological example of these events (e.g.,
those confined to the local tissue microenvironment outside
of circulating serum compartment) are the human granuloma
forming diseases like sarcoidosis and tuberculosis (27). In
these disease states, cells of the innate immune response,
principally macrophages, express the same metabolic machinery
to synthesize 1,25(OH)2D intracellularly when presented with
a CYP27B1 activating signal and with sufficient 25(OH)D in
the extracellular space to serve as substrate for the CPY27B1.
When the extracellular concentration of 25(OH)D falls below the
equivalent of 20 ng·mL1or 50 nM, the intracrine production
of 1,25(OH)2D via the CYP27B1-hydroxylase becomes limiting;
unlike the renal CYP27B1, the enzyme in the macrophage
is highly substrate-drive (28). Taking the human granuloma-
forming, macrophage dominant infectious disease tuberculosis
(TB) as an example, in the face of deficient extracellular
substrate 25(OH)D the macrophage CYP27B1 is unable to
generate enough active 1,25(OH)2D metabolite to effectively
ligand sufficient VDR in that cell to promote expression of
vitamin D-dependent antimicrobial genes (29,30). The end result
is failure of the macrophage to mount an effective autophagy-
related, vesicular killing response to ingested Mycobacterium
Frontiers in Endocrinology | www.frontiersin.org 2October 2019 | Volume 10 | Article 718
50
Chun et al. Free 25D
FIGURE 1 | Vitamin D metabolism and action. Vitamin D metabolites (Left). 7-dehydrocholesterol (7-DHC) is photoconverted to vitamin D3by UV exposure of skin.
CYP2R1 in the liver hydroxylates vitamin D3to 25-hydroxyvitamin D3(25(OH)D). Another hydroxylation to 25(OH)D3by CYP27B1 occurs in the kidney but also in a
number of extra-renal tissues produces the active 1,25-dihydroxyvitamin D3(1,25(OH)2D3). 1,25(OH)2D3is the cognate ligand for the vitamin D receptor (VDR), a
nuclear transcription factor, that directs 1,25(OH)2D3regulated gene transcription. CYP24A1 is responsible for the hydroxylation that yields 24,25-dihydroxyvitamin D
(24,25(OH)2D3) and subsequent catabolism to non-biologically active metabolites. Intracrine mechanism in immune cells (Right). Vitamin D action in immune cells is
reliant upon the intracrine (local) production of active 1,25(OH)2D within the macrophage. Vitamin D status of the host as defined by serum, extracellular 25(OH)D
levels impact immune response, as 25(OH)D is the substrate for CYP27B1. A complex interplay between monokine signaling, that can both be responsive to and
stimulatory of 1,25(OH)2D synthesis, and regulatory signaling among innate and adaptive immune cells is shown.
TABLE 1 | Vitamin D and human health.
Adverse health outcomes significantly associated with low serum
25(OH)D level
Low bone density Obesity
Hip fractures Insulin resistance
Non-vertebral fractures Type 1 diabetes
Heart attack Type 2 diabetes
Hypertension Cancer
Stroke Preterm delivery
Neurocognitive dysfunction Pre-eclampsia
Proximal muscle weakness Inflammation/infection
Autoimmune diseases Multiple sclerosis
Numerous health conditions have been associated with low total serum levels of 25(OH)D
and are listed in the table. Conditions pertaining to bone health are indicated in blue. Table
is based on reviews by Rosen et al. (2) and Rosen and Taylor (4).
tuberculosis (M. tb) (31,32). In vitro this failure can be rescued
in a 25(OH)D concentration-dependent fashion by exchanging
vitamin D deficient human serum with vitamin D sufficient
serum (>30 ng·mL-1 or 75 nM); in other words, rescue of
the macrophage innate immune is achieved by conditioning
activated macrophages ex vivo in serum from the same host after
treatment of the host with vitamin D in vivo (30). Demonstrating
successful rescue from 25(OH)D deficiency in vivo in humans
exposed to or in the very early phases of infection with M. tb.
would support the value of 25(OH)D-driven innate immune
competence in prevention and use as adjunctive therapy early in
the course of this disease (33).
The local antimicrobial capacity of the macrophage is
subject to intracrine, autocrine, and paracrine feedforward
and feedback immune regulatory circuits. This regulatory
network is depicted schematically in the right panel of
Figure 1. Stimulation of the Toll-like receptor signaling pathway
by pathogen-associated molecular pattern (PAMP) molecules
induces expression of CYP27B1 and VDR as well as of monokines
(e.g., IL-15 and IL-1β). In an autocrine mode, these two
monokines act to amplify expression of the CYP27B1 and
1,25(OH)2D-VDR-directed generation of antimicrobial peptides
(34). In a paracrine fashion IL-1ß mobilizes and activates
cells of the adaptive immune response (3537). Activation
of the Th1 subset of “helper” lymphocytes promotes: (1)
production of IFN-γ, the most potent known stimulator of
the macrophage CYP27B1-hydroxylase (38); and (2) induction
of expression of the VDR in adaptive immune response cells
(39,40). When IFN-γ-driven production of 1,25(OH)2D in the
macrophage is robust enough to allow escape of the active
vitamin D metabolite into the local, pericellular inflammatory
microenvironment, this 1,25(OH)2D is sufficient to drive VDR-
dependent gene expression in activated lymphocytes such as
inhibiting proliferation of those lymphocytes. As such, the
predominant paracrine action of 1,25(OH)2D in this setting is
to modify the adaptive immune response (41) and turn down
IFN-γand macrophage CYP27B1 gene expression, preventing a
potential overzealous adaptive (auto)immune response harmful
to the host. Therefore, 25(OH)D “sufficiency” in the serum of
the host appears to be paramount in providing the optimal IFN-
γ-mediated feedback control on 1,25(OH)2D synthesis by the
macrophage and appropriate antimicrobial response to ingested
microbes. For example, failure of this normal feedback control
in disseminated infection with M. tb. may result in escape of
1,25(OH)2D from the local immune microenvironment into
the serum tuberculosis, resulting in a form of endocrine-acting
Frontiers in Endocrinology | www.frontiersin.org 3October 2019 | Volume 10 | Article 718
51
Chun et al. Free 25D
“1,25(OH)2D intoxication” and life-threatening hypercalcemia.
This form of extra-renal 1,25(OH)2D intoxication can occur
in certain granuloma-forming diseases such as tuberculosis
(where pathogen is known) and sarcoidosis (where pathogen
is unknown).
These scenarios indicate that the serum level of
bioavailable 25(OH)D to macrophages is a key determinant
of normal/abnormal physiological control of innate and
adaptive immunity in the host with low serum 25(OH)D
levels. Cell and molecular biology experiments have established
co-existent expression of the CYP27B1 and VDR in the same
cell (Table 2). This observation makes all of these cell-types
potential candidates for intracrine metabolism and action of
1,25(OH)2D generated from 25(OH)D available to that cell from
its extracellular microenvironment in vivo. Beyond tuberculosis
just discussed, many human diseases are associated with “low”
serum 25(OH)D levels compared to matched controls without
the disease (Table 1). In cardiovascular disease, the leading cause
of mortality in the US, mortality is inversely related to serum
25(OH)D <20 ng/ml (42,43), though this is disputed (44). All
of these disease states are associated with altered host immunity.
Perhaps this is the common link to adverse outcomes in this
otherwise diverse set of disorders.
DEFINING VITAMIN D STATUS
The total serum 25(OH)D level (the sum of 25(OH)D that
is bound to carrier proteins and is free in the circulation) is
the currently accepted marker of choice for defining vitamin
D status in any given individual. Its preferential use is based
on (i) analysis of the existing body of clinical research, (ii)
assay advances that confer easy and accurate measurements
and (iii) the fact that it is the most abundant and stable of
the various vitamin D metabolites in serum; 25(OH)D has a
relatively long half-life (15 days) compared to 1,25(OH)2D (15 h).
However, health organizations across the globe differ significantly
in their definition of what level of total circulating 25(OH)D
constitutes sufficiency and deficiency for a “normal” population
without evidence of active bone disease (e.g., osteoporosis,
TABLE 2 | Cell types that express both CYP27B1 and VDR.
Cells co-expressing a functional CYP27B1 and VDR
Macrophage Enterocyte
Dendritic cell Decidual stromal cell
Parathyroid cell Fetal trophoblast
Osteoblast Prostate epithelial cell
Osteoclast Vascular endothelial cell
Keratinocyte Pancreatic βcell
Mammary epithelial cell Renal tubular cell
In contrast to the endocrine action based on kidney production of 1,25(OH)2D that travels
through the general circulation to VDR-possessing target cells, a number of cell types
harbor both CYP27B1 and VDR making intracrine metabolism and action possible. The
red arrow indicates the macrophage, the cell type in which the intracrine metabolism of
25(OH)D to 1,25(OH)2D and 1,25(OH)2D-directed gene expression has been most clearly
demonstrated ex vivo.
hyperparathyroidism, etc.). In North America, the Institute of
Medicine has recommended the value of 20 ng/ml 25(OH)D
(50 nmol/L) for sufficiency (45), whereas the Endocrine Society
recommended for 30 ng/ml (75 nmol/L) (46). The Vitamin
D Council states that individuals should strive for levels above
40 ng/ml (100 nmol/L) (47). In contrast to this, the UK Science
Advisory Council on Nutrition defined vitamin D deficiency as
serum levels of 25(OH)D <10 ng/ml (25 nmol/L), but did not
recommend an optimal level for human health (48). Although the
10 ng/ml level is held by some other European nations, some have
recommended higher levels (49). Investigators in the vitamin D
field have highlighted the limitations of the total 25(OH)D as the
routine biomarker for vitamin D status in various commentary
and review articles (50,51) and discussed other potential markers
and the possible need of a “vitamin D panel” (52,53).
Human data indicates that the threshold for detection of
a relative increase in the serum iPTH at the individual and
population level occurs when the total serum 25(OH)D level
falls below 30 ng/ml (54,55). Thus, PTH offers a measurable
biological consequence of “low” 25(OH)D. However, PTH
levels are not exclusively controled by 25(OH)D as serum
concentration of ionized calcium is sensed at the parathyroid
gland by the calcium-sensing receptor (CaSR). When serum
calcium levels drop, CaSR signal transduction in the parathyroid
yields an increase in PTH production that then enters the general
circulation resulting in PTH’s endocrine effects.
Another reason for the uncertainty concerning the validity of
total 25(OH)D as an exclusive serum marker for vitamin D health
is due to the complex molecular biology and biochemistry of
25(OH)D-associated bioactivities. This is particularly relevant to
the intracrine, paracrine or endocrine conversion of 25(OH)D to
active 1,25(OH)2D, because (i) the local concentrations vitamin
D metabolites outside of the serum compartment cannot be
easily measured in vivo and (ii) the subsequent molecular actions
of 1,25(OH)2D in conjunction with its binding by the VDR
is dependent on diverse mechanisms beyond simple variations
in circulating 25(OH)D. These include: (1) the transport and
target tissue uptake of 25(OH)D; (2) the directed intracellular
transport of 25(OH)D to the inner mitochondrial to CYP27B1
for enzymatic conversion of 25(OH)D to 1,25(OH)2D; (3) export
if 1,25(OH)2D from the mitochondia and binding of 1,25(OH)2D
to VDR; and (4) competing catabolism of 1,25(OH)2D by
the enzyme CYP24A1 also located on the inner mitochondrail
membrane. In this review, we will briefly discuss the molecular
biology and biochemistry behind these processes, with particular
emphasis on the role of free 25(OH)D as a key determinant of
the downstream actions of 1,25(OH)2D, specifically in bone and
mineral health.
MOLECULAR BIOLOGY AND
BIOCHEMISTRY OF VITAMIN D ACTION
1,25(OH)2D is the active vitamin D molecule with 25(OH)D
being its immediate precursor (panel A, Figure 1). It is
1,25(OH)2D that drives vitamin D-regulated gene expression
in target cells. Under normal conditions, the level of serum
Frontiers in Endocrinology | www.frontiersin.org 4October 2019 | Volume 10 | Article 718
52
Chun et al. Free 25D
1,25(OH)2D is tightly regulated within a narrow range (30–
60 pg/ml) at a level that is 1,000X less plentiful than its
25(OH)D precursor. In non-pregnant humans, 1,25(OH)2D
in the serum comes almost exclusively from expression of
CYP27B1 in the kidney. Circulating 1,25(OH)2D’s endocrine
actions are to regulate the serum level of calcium by optimizing
intestinal calcium absorption and/or calcium resorption from
the skeleton (56). Not surprisingly then, when kidney failure
occurs, there is (i) an accompanying decrease in renal CYP27B1
capacity, (ii) a fall in 1,25(OH)2D production and (iii) a
reduction in the serum calcium level. In the opposite case,
when a pathophysiological extra-renal source of CYP27B1
(e.g., the macrophage in granuloma-forming diseases; see
right panel, Figure 1 and Table 1) becomes dysregulated
and dominant, serum 1,25(OH)2D-driven hypercalcemia
and/or hypercalciuria [1,25(OH)2D intoxication] occurs
overriding the calcium-lowering actions of the CaSR in
the parathyroid gland and in the kidney (57). Only in
these two abnormal calcemic states is the serum level of
1,25(OH)2D of the host informative to the clinician in evaluating
a patient.
At the molecular level, 1,25(OH)2D binds VDR with the
highest affinity among all vitamin D metabolites regardless of
whether 1,25(OH)2D is coming from the serum outside the
target cell (endocrine mode), from the local microenvironment
outside of the serum compartment (paracrine mode), or from
the inside of the cell (intracrine mode) (58). In the 1,25(OH)2D-
liganded state the VDR preferentially forms heterodimers
with retinoic acid X receptor (RXR). In the 1,25(OH)2D-
occupied state the VDR and its unliganded RXR partner
heterodimer become transacting complexes binding to specific
cis-acting vitamin D response elements (VDREs) in the genome.
The heterodimer interacts with the transcriptional machinery
resulting in 1,25(OH)2D-regulated (positive or negative) gene
expression and corresponding bioactivities. Due to the three-
dimensional “looping” nature of DNA-protein interactions,
VDRE-like enhancer and repressor motifs can found at
considerable distance from the transcriptional start site of a
vitamin D regulated gene (59,60).
Owing to a rapidly growing skeleton with a high demand for
calcium and phosphate for skeletal mineralization and in the
face of normal VDR and CYP27B1 activity, children suffering
from low 25(OH)D levels and secondary decreases in optimal
intestinal absorption in dietary calcium and phosphate can
develop vitamin D deficient rickets (61). This is commonly
observed in subpopulations of impoverished, dark-skinned
children (i) who require up to 10 times more cutaneous
sunlight exposure that lightly pigmented children to make
the same amount of vitamin D in their skin and (ii) in whom
consumption of natural vitamin D-rich foods (e.g., fish) and
vitamin D supplemented foods is compromised. In nations of
lower income, children usually consume a diet rich in grains;
grains are a rich source of phytates known to chelate ingested
calcium further decreasing intestinal calcium absorption.
Vitamin D deficiency rickets can also be hastened in children
subjected to religious/cultural practices that (e.g., occlusive
garb) that effectively eliminate skin exposure to sunlight. In
vitamin D deficient rickets the serum calcium and phosphate
is low and PTH and 1,25(OH)2D usually elevated for the
subject’s age (62). A low 25(OH)D level (usually <10 ng/ml)
is the distinguishing marker for vitamin D deficient rickets,
distinguishing it from Human Vitamin D Resistant Rickets
(HVDRR; high 1,25(OH)2D) and Pseudo Vitamin D Deficient
Rickets (PDDR; low 1,25(OH)2D) (63). Appropriate dietary
supplementation with calcium and vitamin D to normalize the
serum 25(OH)D level (e.g., >30 ng/ml) alleviates nutritional
rickets (61,64).
Prevention of nutritional rickets in children and osteomalacia
in adults is the primary concern behind the recommendations
on vitamin D intake and serum 25(OH)D level attainment.
Various medical organizations have evaluated the existing body
of research data to support their positions and are exhaustively
reported elsewhere (45,48). However, two key studies are
emblematic of the basis for the determinations. In one line
of evidence, PTH is used as a biomarker of bone health. In
some studies, an inverse association between PTH (declining)
and 25(OH)D (increasing) has been observed (55,65,66). In
one study involving 1,536 post-menopausal women (55) an
inflection point where the decline in serum PTH levels off
was identified at a 25(OH)D level of 30 ng/ml, leading some
authorities in the vitamin D field to call for this to demarcate
the cutoff for vitamin D sufficiency (46). Another often quoted
study involved post-mortem (N=675) determination of
25(OH)D levels and comparisons to histomorphometric analysis
of transiliac crest biopsies (67). From this dataset, the IOM
(45) concluded that at a serum 25(OH)D level of 20 ng/ml and
above, 99% of the normal population (e.g., without a known bone
disease) should have no pathological accumulation of osteoid
(unmineralized portion of bone indicative of rickets in children
and osteomalacia in adults). Interestingly, the original authors
(67) using different criteria to analyze the same data concluded
that 30 ng/ml 25(OH)D as the recommended level for optimal
skeletal health.
TOTAL 25(OH)D OR FREE 25(OH)D?
The measured total 25(OH)D concentration in serum is present
at nearly 1,000-fold higher levels compared to 1,25(OH)2D in
serum and easily and accurately measured from a small amount
of sample (25–50 µl) (68). As such, total 25(OH)D has become
the de facto biomarker of the state of vitamin D deficiency
or sufficiency of the host. However, the majority (>99%) of
serum 25(OH)D is bound (Figure 1, left box) to carrier proteins
(85% to vitamin D binding protein [DBP]; 15% to albumin).
The affinity of DBP for 25(OH)D is 1,000-fold greater than
that of albumin for 25(OH)D (11). Cells that express the cell
surface receptor proteins megalin and cubulin can internalize
the DBP-bound-25(OH)D complex into an endolysosome with
ultimate release of 25(OH)D from DBP into the cell interior
for further metabolism and/or catabolism of 25(OH)D. This
mechanism of endocytosis, intracellular release of 25(OH)D from
acid-hydrolyzed DBP has been most clearly demonstrated at the
luminal membrane of the tubular epithelial cells in the kidney
Frontiers in Endocrinology | www.frontiersin.org 5October 2019 | Volume 10 | Article 718
53
Chun et al. Free 25D
(15). Here internalized 25(OH)D is the substrate used by the
CYP27B1 to form 1,25(OH)2D for endocrine distribution. In
cells not expressing megalin, 25(OH)D entry into its target cell
is proposed to be accomplished by diffusion of the unbound,
free 25(OH)D across the lipid bilayer of the plasma membrane
to the cell interior (15). Thus, the biologically relevant substrate
for some cells is DBP-bound 25(OH)D (essentially equal to
total 25(OH)D) and for others it is the free 25(OH)D or a
related metric of bioavailable 25(OH)D (sum of free 25(OH)D
and albumin-bound-25(OH)D).
Though the serum protein carriers of vitamin D metabolites
are well-characterized, it is now clear that vitamin D metabolites
also reside in non-serum locales such as body fat and inside cells,
suggesting that specific vitamin D binding proteins other than
DBP and albumin may also play a role in vitamin D biology.
Notably intracellular vitamin D binding proteins (IDBPs) were
identified as a result of investigations into apparent vitamin D
resistance in New World Primates [NWP; reviewed in Adams
et al. (69,70)]. The high serum levels of steroid hormones
in general and vitamin D in particular, relative to Old World
Primates (OWP), was shown to be associated with a form
of target tissue insensitivity to 1,25(OH)2D (71). The first
indications of a functional role for IDBPs was the observed
diminished ability of NWP cells to effectively upregulate VDR-
target genes, like the CYP24A1, despite having comparable
amounts of VDR (72,73) and VDR functionality (74). Using
cell extracts from NWP and OWP in VDR-1,25(OH)2D
radiolabel binding assays, a protein in NWP cell extracts was
observed to prevent VDR-1,25(OH)2D binding when mixed
with OWP extracts. This inhibition of binding was abolished
after trypsin digestion or heat denaturation (75). Upon further
characterization, IDBP was found to (i) bind 25(OH)D as well as
other steroid hormones (76) and (ii) be part of the heat shock
protein 70 family (77,78). With cDNA constructs for IDBP
obtained, transient and stable overexpression in vitro in tissue
culture studies revealed that IDBP could increase 1,25(OH)2D
synthesis, possibly by chaperoned delivery of 25(OH)D to the
CYP27B1 (79). The ATPase domain of IDBP was essential to this
function (80,81), with the BCL2-associated athanogene 1 (BAG1)
serving as an IDBP co-chaperone (82).
Measurement of the free, unbound form of 25(OH)D in
serum is challenging, because its levels are low (4–8 pg/ml
range) and has historically required radioactive tracers of
25(OH)D with equilibrium or centrifugal dialysis methods
that are cumbersome and impractical for use in clinical
laboratory testing services (10,11). Free and bioavailable
25(OH)D levels in the serum can be mathematically calculated
using equations that incorporate the binding affinity of DBP
for 25(OH)D and the concentration of DBP in the serum
(11,83). This approach of calculating the free, biologically active
fraction of 25(OH)D has also been taken with testosterone
(84) and thyroid hormone (85). Unfortunately, for free
25(OH)D, it was discovered that one commonly used DBP
ELISA kit used to calculate for free 25(OH)D had differential
sensitivities to the common phenotypic variants of DBP
(discussed later in this review) that resulted in inaccurate
measurement of DBP concentrations in some samples
TABLE 3 | Hormone binding proteins found in human serum.
Binding protein Metabolites Percent free
DBP
4.5–5.5 µMa
Total 25(OH)D
25–75 nMa
Free 25(OH)D
5–20 pMa
0.02
Total 1,25(OH)2D
50–198 pMb
Free 1,25(OH)2D
325–525 fMc
0.5
TBG
241–722 nMb
Total T4
58–154 nMb
Free T4
11–23 pMb
0.02
Total T3
1.1–2.8 nMb
Free T3
3.0–6.8 pMb
0.3
SHBG
16.5–55.9 nMb
(male)
Male total T
9.2–31.8 nMb
Male free T
30–87 pMb
0.3
Male total E2
28–156 pMb
Male free E2
<1.7 pMd
1
SHBG
24.6–122.0 nMb
(female)
Female total T
0.3–1.7 nMb
Female free T
<15 pMb
0.9
Female total E2
46–609 pMb
Female free E2
1.6–18.5 pMd
3
aNielson et al. (88).
bhttps://www.labcorp.com/test-menu/ search.
cBikle et al. (89).
dhttps://www.questdiagnostics.com/testcenter/TestCenterHome.action.
Vitamin D binding protein (DBP), thyroxine binding globulin (TBG), and sex hormone
binding globulin (SHBG) are tabulated with their serum concentration levels. The
respective metabolite concentrations total, free and percent free are also presented.
Albumin can bind all the hormones listed and transthyretin can bind T4 albeit at lower
affinity compared to their primary carrier proteins. The bioavailable concept has been
applied to these hormones and comprise the sum of albumin-bound-hormone and
free hormone.
leading to inaccurate calculated free 25(OH)D levels in
those serum samples.
Direct measurement of the various free hormones of clinical
importance have been developed (86,87). However, this aspect
of clinical chemistry remains challenging due to (i) the low
concentration of these metabolites in the serum (Table 3) and (ii)
variability of serum composition (e.g., lipids and other potentially
interfering molecules) between patients that can introduce error
into the measured value. Recently, an ELISA based method for
detection of free 25(OH)D has been developed (90) and has been
utilized in a number of clinical studies that we will summarize in
the clinical studies portion of this review. Most recently, a high-
throughput method to measure bioavailable 25(OH)D has been
developed (91). However, procedures to ensure accuracy and
precision across a wide range of sample conditions both for both
of these assays have not been yet developed; as a consequence,
these assays are deemed for research use only.
MOLECULAR BIOLOGY AND
BIOCHEMISTRY OF DBP
The vitamin D binding protein (DBP) is a multi-functional
protein also known as Group Specific Component (GC)
[reviewed in Chun (92) and Delanghe et al. (93)]. DBP is
a member of the albumin superfamily of proteins. DBP is a
moderately abundant protein (5µM in humans) in serum of
Frontiers in Endocrinology | www.frontiersin.org 6October 2019 | Volume 10 | Article 718
54
Chun et al. Free 25D
TABLE 4 | Molecular biology of most common DBP polymorphisms.
SNP name GC name Codon variant Amino acid variant
rs4588 GC1 ACG Thr-436
GC2 AAG Lys-436
rs7041 GC1F GAT Asp-432
GC1S GAG Glu-432
Two single nucleotide polymorphisms (SNP) account for three of the major forms of DBP
(originally known as GC-globulin). Table also includes the specific codon and amino acid
variation that define these variants.
vertebrates. In humans the GC/DBP 13 exon gene is located at
4q13.3. Its gene product, DBP, is highly expressed in the liver and
exported into the circulatory system. DBP contains 474 amino
acids of which the 16 N-terminal amino acids function as a
signal peptide. DBP can be glycosylated (94) to varying degrees
depending on genotype. The glycosylation pattern has been
suggested to be structural basis for the macrophage activating
factor activity of DBP (95,96).
DBP’s most well-characterized role is that of a carrier protein
for vitamin D metabolites. Its rank order avidity for the
vitamin D and it metabolites are (10,11,89,97) as follows:
24,25(OH)2D>25(OH)D>1,25(OH)2D>vitamin D. DBP binds
vitamin D3 and its metabolites with greater affinity than vitamin
D2 and it metabolites (97). DBP can also bind actin (98). This
biological action is suggested to be that of scavenging of exposed
actin, preventing overzealous extracellular polymerization after
tissue injury (99). DBP can also bind circulating fatty acids (100)
and C5a des Arg, the latter of which enhances complement
activation (101).
DBP migrates at 52–59 kDa in electrophoretic gels (102).
It was variations in DBP mobility in isoelectric focusing (IEF)
gels that garnered initial research attention prior to its function
being determined. One banding pattern was termed GC-1F
(faster), another GC-1S (slower), and still another GC2 (103).
GC2 migrated less rapidly toward the anode compared to either
GC1 forms. The different forms, due to single amino acid
differences (Table 4), were (i) used to determine allelic frequency
for samples worldwide (104) and (ii) found to associate with the
racial background of the source human serum. In the Kamboh
study, black subjects were more likely to have the GC1F forms
(67–79% of alleles in USA blacks) while white subjects more
frequently yielded the GC1S pattern (49–57% of alleles in USA
whites). The GC2 allele was observed were more frequently seen
in white subject samples (21–31% in USA whites) compared to
blacks (8–13% in USA blacks). These three classic forms (GC1F,
GC1S, and GC2) account for the vast majority of the variation
DBP across human populations. Since these early studies, many
other SNPs (105) have been found in GC of which a small
percentage encodes a missense mutation that changes the amino
acid code.
Affinity Differences and DBP Genotype
The biological significance of DBP’s different allelic forms is
unclear. Conceptually, if the different genotypic forms of DBP
had different affinities for vitamin D metabolites, then the
levels of those metabolites could be influenced by the genotype.
However, in terms of experimentally measured differences in
affinity for vitamin D metabolites, one study reported large
differences (106) but three others did not (107109). Media
supplemented with serum that contained different alleles of
DBP had differing impacts on assessed immunological readouts
consistent with affinity differences in three in vitro studies
(110112). The mechanism for the observed differences were
not investigated in those studies; thus, non-affinity dependent
mechanisms cannot be ruled out. Using an ELISA based assay to
detect free 25(OH)D, some small differences in percent free were
detected between the genotypes (113). However, these differences
were smaller in magnitude than the differences anticipated
if the report of large affinity difference among genotype
was accurate.
DBP Serum Concentration and DBP
Genotype
The other mechanism by which DBP genotype could impact
free 25(OH)D levels is through differentials in the genotype-
dependent DBP concentration. It is on this basis that Powe
et al. (114) proposed how black Americans, with lower total
serum 25(OH)D levels and substantially lower DBP levels
compared to white Americans, could ultimately have similar free
25(OH)D levels. However, it was eventually determined that the
monoclonal antibody-based ELISA used in their study was less
sensitive to the GC1F DBP, the form most frequently found in
blacks (88,115). This resulted in an underestimation of serum
DBP levels in black subjects and a consequent overestimation
of the calculated free and bioavailable serum 25(OH)D levels in
their serum. Nonetheless, there is evidence of some genotypic
effect on DBP concentrations. One early study (116) of Danish
women found DBP concentration ranked according to the
presence of GC1 alleles (GC1-GC1 >GC1-GC2 >GC2-GC2).
Consistent with those findings was a study in a population male
subjects with modest racial diversity where GC2/GC2 subjects
exhibited the lowest serum DBP concentration (88).
In another recent study utilizing serum samples from both
women and men with modest racial diversity, the presence of the
GC2 allele in one of three allelic combinations resulted in lower
DBP levels compared to the three allelic combinations without
GC2 (113). Currently, it is thought that DBP’s role in determining
free 25(OH)D levels is largely through its concentration [i.e.,
for a fixed total 25(OH)D, more DBP yields less free 25(OH)D]
and perhaps to a small degree by genotype. As such, in most
cases total 25(OH)D and free 25(OH)D are highly correlated.
However, there are some conditions where DBP levels diverge
from typical levels. For instance, patients with liver disease
have lower levels of DBP while pregnant women have higher
DBP levels (89,117) resulting in higher free 25(OH)D levels
in those with lower DBP concentrations. In the more extreme
case of patients before [lower DBP, lower 25(OH)D] and after
[higher DBP, higher 25(OH)D] liver transplants, the percentage
of free 25(OH)D relative to total 25(OH)D is higher before
transplantation. Recently, a case report described a subject with
Frontiers in Endocrinology | www.frontiersin.org 7October 2019 | Volume 10 | Article 718
55
Chun et al. Free 25D
a homozygous deletion of DBP (118). Compared to unaffected
and heterozygous siblings, the affected patient had no DBP and
very low serum levels of 25(OH)D and 1,25(OH)2D. Despite the
extremely low serum levels of vitamin D metabolites, the subject
had normal calcium balance with only relatively small alterations
in bone health and mineral metabolism. This case report is the
strongest evidence to date that the total serum 25(OH)D and
1,25(OH)2D level can be disconnected from “normal” vitamin D
status. One explanation is that albumin, or some other chaperone
in the serum, assumes the role of delivering 25(OH)D and
maintaining viable levels of free 25(OH)D and 1,25(OH)2D,
as was measured in this subject, to target tissues for further
metabolism and action.
ANIMAL STUDIES OF FREE 25(OH)D AND
BONE
Dbp heterozygous and homozygous knockouts in mice do
not have any obvious bone phenotype relative to wild type
(119). However, with the loss of a single copy of Dbp the
levels of circulating total serum 25(OH)D and 1,25(OH)2D are
diminished and when both copies were absent, the levels of serum
25(OH)D and 1,25(OH)2D were extremely low, phenocopying
the DBP/subject just described above. Interestingly, neither
DBP+/or DBP/mice exhibited any skeletal abnormalities
or problems with calcium and phosphate balance (119). These
findings support the hypothesis that free hormone levels alone
are adequate for sustaining skeletal health. Consistent with this
was another study with Dbp homozygous knockouts where the
levels of 1,25(OH)2D was measured in the intestinal tissues (120).
Even though the double knockout animals had very low total
serum 1,25(OH)2D levels compared to wild type, the levels of
1,25(OH)2D measured in the intestinal tissues were very similar.
These findings suggest that trace amounts of free metabolites
and/or enhanced local conversion of 25(OH)D to 1,25(OH)2D
are sufficient as long as sufficient amounts substrate 25(OH)D
are available to the host. The fact that mice with no DBP are still
viable suggests that albumin, though having a much lower affinity
for vitamin D metabolites, could serve as the carrier protein
in place of DBP. When Dbp null mice were raised on vitamin
D3-free diets, they developed secondary hyperparathyroidism
and bone mineralization defects much more rapidly than paired
wild-type mice (119). This result indicated that DBP’s function
is to maintain a stable reservoir of circulating extracellular
vitamin D metabolites. Owing to its higher affinity for vitamin
D metabolites, DBP is more effective in this role than albumin,
despite both DBP and albumin being filtered into the urine and
reclaimed by megalin.
Another test of the biological impact of free 25(OH)D in mice
(121) was based on the difference in affinity of DBP for D2 or
ergocalciferol (vitamin D found in fungi) vs. D3 or cholecalciferol
(vitamin D found in animals) forms of vitamin D metabolites.
Mice raised on diets containing exclusively D2 or D3 would result
in the mice having only 25(OH)D2 and 25(OH)D3 circulating in
their serum. Since DBP affinity for D2 forms is lower relative to
D3 forms, the free 25(OH)D2 levels were expected to be higher
in animals raised on D2 diets. In this study, mice were placed
on D2 or D3 diets (1,000 IU/kg) beginning at week 3 and tested
at week 8 and week 16 for 25(OH)D3 and 25(OH)D2 serum
levels and bone phenotype by histomorphometry. These mice
had similar total 25(OH)D levels at week 8 (26.6 ±1.9 ng/ml
25(OH)D2 vs. 28.3 ±2.0 ng/ml 25(OH)D3) and at week 16 (33.3
±4.4 vs. 31.7 ±2.1 ng/ml). However, as anticipated, they differed
in their free 25(OH)D levels with free 25(OH)D2 greater than
free 25(OH)D3 at week 8 (16.8 ±0.65 vs. 8.4 ±0.63 pg/ml,
P<0.001) and at week 16 (17.4 ±0.43 vs. 8.4 ±0.44, P<
0.001). Histomorphometric analysis of their bones detected that
at week 8, the D2 fed mice had significantly higher osteoclast
surface/bone surface, eroded surface/bone surface, and mineral
apposition rate (high bone turnover) compared with mice raised
on the D3 diets. Additionally, osteoblast surface/bone surface, an
index of bone formation, was higher in week 8 D2 in females
only. The reason underpinning this sexual dimorphism in bone
formation rates remains unknown. The bone phenotype at week
16 revealed significantly higher bone volume/total volume and
trabecular number in the D2 mice relative to the D3 mice.
Taken together, despite similar total serum 25(OH)D levels, bone
phenotype differences were observed in association with different
free 25(OH)D levels (higher in D2 mice) suggesting the relevance
of free 25(OH)D to bone health.
To the best of our knowledge, there have been no
studies on the effects of free 25(OH)D and the bone health
of animals besides the studies in mice summarized above.
However, the existence of nocturnal bats that roost in dark
locations (122) could be informative. These bats have very
low total serum 25(OH)D (<5 ng/ml) as their normal state
suggesting that “deficient” (by human standards) total 25(OH)D
levels presumably still yields adequate free 25(OH)D levels to
sustain normal bone and mineral homeostasis for this species.
Interestingly, these bats are fully capable of attaining higher
25(OH)D levels when housed in conditions that expose them to
sunlight. Perhaps these animals have lower DBP concentrations
or metabolite affinities for DBP to compensate for their naturally
low total serum 25(OH)D levels. A test of these animal’s
serum with the free 25(OH)D assay could be informative.
Other possible adaptations that permit compensation to the
very low circulating levels of 25(OH)D include: (1) a highly
efficient CYP27B1; (2) hyper-sensitive VDR to ligand; (3)
heightened transactivation potential of VDR-interacting co-
activators; and/or (4) diminished functional activity of the
CYP24A1 catabolic machinery. There is one study comparing
DBP affinity for 25-hydroxyvitamin D among several animals;
these investigators found the DBP from rat and cattle exhibited
higher affinity to 25(OH)D compared to horse and rhesus
monkey with humans having the lowest affinity of species tested
(123). What the levels of free 25(OH)D are and its importance to
these species have not yet been examined.
HUMAN STUDIES OF FREE 25(OH)D
Revitalized interest on the impact of vitamin D on human
health beyond bone was spurred by studies that investigated
Frontiers in Endocrinology | www.frontiersin.org 8October 2019 | Volume 10 | Article 718
56
Chun et al. Free 25D
vitamin D’s regulatory role in the adaptive and innate immune
system (26,29,124). Three in vitro studies have demonstrated
that decreasing free 25(OH)D by increasing DBP in the culture
media diminished immune functions of adherent monocytes
(110), dendritic cells (111), and T-lymphocytes (112). Because
of these findings in vitro, the parameter of free 25(OH)D
began to receive greater interest in clinical research. In
non-bone health association studies, the results have been
mixed. In some studies free serum 25(OH)D levels were
inversely associated with coronary artery disease (125), pediatric
inflammatory bowel disease (126) and ulcerative colitis (127),
insulin sensitivity (128), reduction in lipid markers in statin
patients (129), and acromegaly (130). However, in some other
studies free 25(OH)D was inferior in asthma (131) and no
better than total 25(OH)D for colorectal cancer in African-
Americans (132).
Concerning bone and mineral health, a 2011 report (133)
was the first to utilize free 25(OH)D and the related concept
bioavailable 25(OH)D (sum of free 25(OH)D and albumin-
bound 25(OH)D) for analytical purposes. They found that these
two metrics of serum vitamin D status were more closely and
directly associated with BMD in the individual than total serum
25(OH)D. This group then followed up with another report
(134) that showed that compared to measures of total 25(OH)D
bioavailable 25(OH)D had a significant direct association with
serum calcium (corrected for the serum albumin level) and a
significant inverse association with PTH. These investigators
suggested that this could address the long-standing paradox
of how black Americans with lower total serum 25(OH)D
levels have higher BMD and similar PTH levels compared
to white Americans. In additional work, they measured DBP
serum concentrations in black Americans and found them
to be lower than those in white Americans. They concluded
that the resultant higher bioavailable 25(OH)D may account
for the nominal differences in BMD and serum PTH (114)
despite what appeared to be sub-optimal total serum 25(OH)D
levels; unfortunately, a high-throughput assay to directly measure
free 25(OH)D was not available at that time. Thus, in these
reports, measured DBP, albumin and total 25(OH)D values were
input into mathematical equations (11,84) to calculate values
for free 25(OH)D and bioavailable 25(OH)D. Disappointingly,
one of the popular ELISA kits for DBP quantitation at the
time relied upon a monoclonal antibody that turned out to
possess reduced sensitivity to the DBP polymorphism (GC1F)
most frequently found in black Americans (88,115). Because
of this characteristic of the ELISA, the DBP concentration
was under-reported for these subjects leading to an over-
estimation of the calculated free and bioavailable 25(OH)D. The
manufacturer of this monoclonal ELISA has re-designed their
ELISA and released a new version in January 2017 addressing
this problem (135). Even with these complications, these early
studies sparked interest in examining bone and mineral health
by markers of vitamin D status other than the traditional total
serum 25(OH)D.
Investigators have continued to use calculated free and
bioavailable 25(OH)D in serum in association studies. In light
of the difficulties with the monoclonal antibody-based ELISA
for DBP, immunological methods based on polyclonal antibodies
(less influenced by DBP polymorphisms) and techniques
independent of antibodies entirely (mass spectrometry) can
be employed to measure DBP for calculation of bioavailable
25(OH)D in serum (88,115). Additionally, an ELISA method
has been developed that measures free 25(OH)D directly (90)
such that some investigators use this assay exclusively in their
studies though many also include data from calculated free
and/or bioavailable 25(OH)D.
HUMAN CLINICAL STUDIES OF FREE
25(OH)D AND BONE
This section surveys the recent literature examining whether free
(directly measured or calculated using the polyclonal DBP assay)
vs. total 25(OH)D is more consistently associated with various
measures of bone health, including intestinal calcium absorption,
parathyroid hormone secretion, and bone mineral density.
Intestinal Calcium Absorption
To our knowledge, there has only been one study to date
examining the relation between free and total 25(OH)D with
intestinal calcium absorption. Aloia et al. randomized 71 adults
to receive either placebo, 800, 2,000, or 4,000 IU/days of
vitamin D3 over 8 weeks. At both baseline and follow-up,
neither free nor total 25(OH)D nor 1,25(OH)2D in the serum
was associated with intestinal calcium absorption efficiency
(136). This supports the concept that VDR-directed increases in
intestinal calcium absorption are controlled locally, outside of the
serum compartment.
Parathyroid Hormone
Multiple investigators have assessed whether free vs. total
25(OH)D is more strongly correlated with the serum PTH level,
with results being inconsistent. For example, in an analysis of
155 subjects that included 24 cirrhotics and 20 pregnant women,
Schwartz et al. reported that both free and total serum 25(OH)D
were similarly, inversely correlated with the serum PTH (117).
Similar findings have been reported in: (1) healthy pre- (137)
and postmenopausal women (136); (2) individuals with obesity
(138); (3) patients cirrhosis of the liver (139); (4) children (2–18
y/o) in Spain (140); (5) blacks and whites in a supplementation
study (placebo, 2,000, 4,000 IU/days for 16 weeks) (141); (6)
a RCT of prediabetics (142); and (7) pregnant white women
in Germany (143). While the above studies reported similar
correlations between serum free and total 25(OH)D with serum
PTH, others have favored free 25(OH)D. For example, Schwartz
et al. conducted a 16-weeks trial in which 81 older women
and men received vitamin D3 at doses of 800, 2,000, or 5,000
IU/days or 50,000 IU/weeks. At the end of the study, free, but
not total serum 25(OH)D was inversely associated with the
serum PTH; however, free 25(OH)D explained only a small
amount of the variability in PTH [R2=0.08; (144)]. In two,
smaller trials – one in which 38 participants received 500,000
IU of vitamin D2 or D3 over 10 weeks (145) and another in
which 35 participants received 2,400 IU/day of vitamin D3 or 20
mcg/day of 25-hydroxyvitamin D3 (146)—Shieh et al. reported
Frontiers in Endocrinology | www.frontiersin.org 9October 2019 | Volume 10 | Article 718
57
Chun et al. Free 25D
that longitudinal increase in free 25(OH)D was significantly
associated with concurrent decrease in serum PTH during the
first 8–10 weeks of supplementation (when 25(OH)D levels
change most rapidly), whereas increase in total 25(OH)D was
not. In adults with primary hyperparathyroidism, Wang et al.,
similarly found that the free serum 25(OH)D was inversely
correlated with circulating PTH levels, but total 25(OH)D was
not (147). Further complicating the picture are studies favoring
total 25(OH)D over free 25(OH)D. In a cohort of Hungarian
adults assessed at the end of winter total, but not free 25(OH)D,
was inversely correlated with PTH (148). In a study of UK whites
and south Asians, total, but not free 25(OH)D, was inversely
correlated with PTH (149). In a study of pregnant adolescents
(13–18 y/o), the inverse association of PTH with free 25(OH)D
was weaker than that observed total 25(OH)D (150).
Bone Mineral Density
While a change in the serum PTH level is the outcome that has
been most frequently tested in relation to free vs. total 25(OH)D,
some cross-sectional studies have examined bone mineral density
(BMD) as well. As was the case with changes in the serum
PTH, results with BMD have been inconsistent. For example,
Jemielita et al. reported that in 304 adults, neither total nor free
serum 25(OH)D at a single point in time was associated with
BMD assessed by DXA, or peripheral quantitative CT (151). In
contrast, in a cross-sectional analysis comparing the correlations
between free vs. total serum 25(OH)D with BMD and composite
indices of femoral neck strength, Alwan reported that higher free
25(OH)D levels were correlated with greater BMD (lumbar spine,
femoral neck, total hip), and femoral neck strength (r=0.24–
0.34, p<0.05), but total 25(OH)D was not (152). On the flip
side, Michaelsson et al. reported that in women from Sweden
(mean age 68 years) higher total, but not free, serum 25(OH)D
was associated with greater BMD (153).
CHALLENGES AND PROSPECTS
Undoubtedly, further clinical studies should be conducted using
bone/mineral outcomes as well as non-skeletal health readouts
to assess the utility of free 25(OH)D. As described in the
above section pertaining to bone and mineral health, human
studies comparing whether free vs. total serum25(OH)D is
more frequently associated with intestinal calcium absorption,
parathyroid hormone secretion, or bone mineral density have
yielded inconsistent results ranging from no difference, to those
favoring either free, or total 25(OH)D. We propose that there
are two major challenges that contribute to these inconsistencies.
First, is the lack of a specific “readout” of vitamin D bioactivity.
While circulating PTH levels are influenced by vitamin D status
(154156), it is also regulated by the calcium sensing receptor.
Additionally, attempts to associate BMD with vitamin D status
are complicated by the fact that BMD in adults is principally
determined by attained peak bone mass that is partly dependent
on vitamin D status (157). Thus, it is difficult to discern the
relative importance of free vs. 25(OH)D in human cross-sectional
studies using these parameters. As such, even when studies
report statistically significant correlations, the r values tend
to fall in the range (0.3–0.1 and 0.1–0.3) that are deemed
“weak relationship” by statisticians. Second, many human studies
do not employ subjects who are 25(OH)D deficient. These
subjects would have the most to gain physiologically from
treatment to return 25(OH)D in the serum to normal. Ideally,
supplementation studies must include subjects that are clearly
at insufficient levels and then have it demonstrably shown that
their levels are raised into the sufficient range. It is through
longer-term longitudinal analyses of intra-individual changes in
serum total and free 25(OH)D after an aggressive vitamin D
supplementation regimen that would have a greater likelihood to
detect any associations.
Lastly, there is currently only one method of direct
measurement of free 25(OH)D (90) with reasonable throughput.
Though very promising, this assay is based on antibody
interaction with 25(OH)D and needs further validation on
the wide variety of sample quality (i.e., time from collection
to testing, temperature of storage, variability of potentially
interfering serum components among patients, etc.) encountered
in clinical laboratory practice (158). In the future, perhaps a mass
spectrometry-based method could be developed as is occurring in
the measurement of other free hormones such as estradiol (159),
thyroid hormone (160), and testosterone (161).
AUTHOR CONTRIBUTIONS
JA developed the overall organization of and approved this
publication. JA, RC, AS, CG, and MH wrote sections of this
review. VY and JW participated in copy editing.
FUNDING
This work was supported by NIH 5R01AR063910-05.
REFERENCES
1. Rosen CJ, Abrams SA, Aloia JF, Brannon PM, Clinton SK, Durazo-
Arvizu RA, et al. IOM committee members respond to Endocrine
Society vitamin D guideline. J Clin Endocrinol Metab. (2012) 97:1146–52.
doi: 10.1210/jc.2011-2218
2. Rosen CJ, Adams JS, Bikle DD, Black DM, Demay MB, Manson JE,
et al. The nonskeletal effects of vitamin D: an Endocrine Society
scientific statement. Endocr Rev. (2012) 33:456–92. doi: 10.1210/er.20
12-1000
3. Christakos S, Hewison M, Gardner DG, Wagner CL, Sergeev IN, Rutten
E, et al. Vitamin D: beyond bone. Ann N Y Acad Sci. (2013) 1287:45–58.
doi: 10.1111/nyas.12129
4. Rosen CJ, Taylor CL. Common misconceptions about vitamin D–
implications for clinicians. Nat Rev Endocrinol. (2013) 9:434–8.
doi: 10.1038/nrendo.2013.75
5. Bouillon R, Marcocci C, Carmeliet G, Bikle D, White JH, Dawson-
Hughes B, et al. Skeletal and extra-skeletal actions of vitamin D: current
evidence and outstanding questions. Endocr Rev. (2018) 40:1109–51.
doi: 10.1210/er.2018-00126
Frontiers in Endocrinology | www.frontiersin.org 10 October 2019 | Volume 10 | Article 718
58
Chun et al. Free 25D
6. Holick MF, Richtand NM, McNeill SC, Holick SA, Frommer JE, Henley
JW, et al. Isolation and identification of previtamin D3 from the skin
of rats exposed to ultraviolet irradiation. Biochemistry. (1979) 18:1003–8.
doi: 10.1021/bi00573a011
7. Holick MF, MacLaughlin JA, Clark MB, Holick SA, Potts JT Jr, Anderson RR,
et al. Photosynthesis of previtamin D3 in human skin and the physiologic
consequences. Science. (1980) 210:203–5. doi: 10.1126/science.6251551
8. Holick MF, MacLaughlin JA, Doppelt SH. Regulation of cutaneous
previtamin D3 photosynthesis in man: skin pigment is not an essential
regulator. Science. (1981) 211:590–3. doi: 10.1126/science.6256855
9. Chen TC, Chimeh F, Lu Z, Mathieu J, Person KS, Zhang A, et al. Factors
that influence the cutaneous synthesis and dietary sources of vitamin D. Arch
Biochem Biophys. (2007) 460:213–7. doi: 10.1016/j.abb.2006.12.017
10. Bikle DD, Siiteri PK, Ryzen E, Haddad JG. Serum protein binding
of 1,25-dihydroxyvitamin D: a reevaluation by direct measurement
of free metabolite levels. J Clin Endocrinol Metab. (1985) 61:969–75.
doi: 10.1210/jcem-61-5-969
11. 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)
63:954–9. doi: 10.1210/jcem-63-4-954
12. Cheng JB, Levine MA, Bell NH, Mangelsdorf DJ, Russell DW. Genetic
evidence that the human CYP2R1 enzyme is a key vitamin D 25-hydroxylase.
Proc Natl Acad Sci USA. (2004) 101:7711–5. doi: 10.1073/pnas.0402490101
13. Zhu JG, Ochalek JT, Kaufmann M, Jones G, Deluca HF. CYP2R1 is a major,
but not exclusive, contributor to 25-hydroxyvitamin D production in vivo.
Proc Natl Acad Sci USA. (2013) 110:15650–5. doi: 10.1073/pnas.1315006110
14. Avioli LV, Lee SW, McDonald JE, Lund J, DeLuca HF. Metabolism of vitamin
D3-3H in human subjects: distribution in blood, bile, feces, and urine. J Clin
Invest. (1967) 46:983–92. doi: 10.1172/JCI105605
15. Nykjaer A, Dragun D, Walther D, Vorum H, Jacobsen C, Herz
J, et al. An endocytic pathway essential for renal uptake and
activation of the steroid 25-(OH) vitamin D3. Cell. (1999) 96:507–15.
doi: 10.1016/S0092-8674(00)80655-8
16. Jones G, Prosser DE, Kaufmann M. 25-Hydroxyvitamin D-24-hydroxylase
(CYP24A1): its important role in the degradation of vitamin D. Arch Biochem
Biophys. (2012) 523:9–18. doi: 10.1016/j.abb.2011.11.003
17. Kremer R, Goltzman D. Parathyroid hormone stimulates mammalian renal
25-hydroxyvitamin D3-1 alpha-hydroxylase in vitro.Endocrinology. (1982)
110:294–6. doi: 10.1210/endo-110-1-294
18. Murayama A, Takeyama K, Kitanaka S, Kodera Y, Hosoya T, Kato S.
The promoter of the human 25-hydroxyvitamin D3 1 alpha-hydroxylase
gene confers positive and negative responsiveness to PTH, calcitonin,
and 1 alpha,25(OH)2D3. Biochem Biophys Res Commun. (1998) 249:11–6.
doi: 10.1006/bbrc.1998.9098
19. Tanaka Y, DeLuca HF. Measurement of mammalian 25-hydroxyvitamin D3
24R-and 1 alpha-hydroxylase. Proc Natl Acad Sci USA. (1981) 78:196–9.
doi: 10.1073/pnas.78.1.196
20. Zierold C, Mings JA, DeLuca HF. Parathyroid hormone regulates 25-
hydroxyvitamin D(3)-24-hydroxylase mRNA by altering its stability. Proc
Natl Acad Sci USA. (2001) 98:13572–6. doi: 10.1073/pnas.241516798
21. Shimada T, Kakitani M, Yamazaki Y, Hasegawa H, Takeuchi Y, Fujita T,
et al. Targeted ablation of Fgf23 demonstrates an essential physiological role
of FGF23 in phosphate and vitamin D metabolism. J Clin Invest. (2004)
113:561–8. doi: 10.1172/JCI19081
22. Shimada T, Hasegawa H, Yamazaki Y, Muto T, Hino R, TakeuchiY, et al. FGF-
23 is a potent regulator of vitamin D metabolism and phosphate homeostasis.
J Bone Miner Res. (2004) 19:429–35. doi: 10.1359/JBMR.0301264
23. Haussler MR, Whitfield GK, Kaneko I, Forster R, Saini R, Hsieh JC,
et al. The role of vitamin D in the FGF23, klotho, and phosphate
bone-kidney endocrine axis. Rev Endocr Metab Disord. (2012) 13:57–69.
doi: 10.1007/s11154-011-9199-8
24. Haussler MR, Whitfield GK, Kaneko I, Haussler CA, Hsieh D, Hsieh JC,
et al. Molecular mechanisms of vitamin D action. Calcif Tissue Int. (2013)
92:77–98. doi: 10.1007/s00223-012-9619-0
25. Holick MF. Vitamin D deficiency. N Engl J Med. (2007) 357:266–81.
doi: 10.1056/NEJMra070553
26. Hewison M. Vitamin D and immune function: autocrine, paracrine
or endocrine? Scand J Clin Lab Invest Suppl. (2012) 243:92–102.
doi: 10.3109/00365513.2012.682862
27. Fuss M, Pepersack T, Gillet C, Karmali R, Corvilain J. Calcium and vitamin
D metabolism in granulomatous diseases. Clin Rheumatol. (1992) 11:28–36.
doi: 10.1007/BF02207080
28. Adams JS, Hewison M. Extrarenal expression of the 25-hydroxyvitamin
D-1-hydroxylase. Arch Biochem Biophys. (2012) 523:95–102.
doi: 10.1016/j.abb.2012.02.016
29. Liu PT, Stenger S, Li H, Wenzel L, Tan BH, Krutzik SR, et al. Toll-like
receptor triggering of a vitamin D-mediated human antimicrobial response.
Science. (2006) 311:1770–3. doi: 10.1126/science.1123933
30. Adams JS, Ren S, Liu PT, Chun RF, Lagishetty V, Gombart AF, et al. Vitamin
d-directed rheostatic regulation of monocyte antibacterial responses. J
Immunol. (2009) 182:4289–95. doi: 10.4049/jimmunol.0803736
31. Fabri M, Stenger S, Shin DM, Yuk JM, Liu PT, Realegeno S, et al.
Vitamin D is required for IFN-gamma-mediated antimicrobial
activity of human macrophages. Sci Transl Med. (2011) 3:104ra102.
doi: 10.1126/scitranslmed.3003045
32. Adams JS, Ramin J, Rafison B, Windon C, Windon A, Liu PT. Redefining
human vitamin D sufficiency: back to the basics. Bone Res. (2013) 1:2–10.
doi: 10.4248/BR201301002
33. Jolliffe DA, Ganmaa D, Wejse C, Raqib R, Haq MA, Salahuddin
N, et al. Adjunctive vitamin D in tuberculosis treatment: meta-
analysis of individual participant data. Eur Respir J. (2019) 53:1802003.
doi: 10.1183/13993003.02003-2018
34. Krutzik SR, Hewison M, Liu PT, Robles JA, Stenger S, Adams JS, et al.
IL-15 links TLR2/1-induced macrophage differentiation to the vitamin
D-dependent antimicrobial pathway. J Immunol. (2008) 181:7115–20.
doi: 10.4049/jimmunol.181.10.7115
35. Dinarello CA. The interleukin-1 family: 10 years of discovery. FASEB J.
(1994) 8:1314–25. doi: 10.1096/fasebj.8.15.8001745
36. Oppenheim JJ, Biragyn A, Kwak LW, Yang D. Roles of antimicrobial peptides
such as defensins in innate and adaptive immunity. Ann Rheum Dis. (2003)
62:ii17–21. doi: 10.1136/ard.62.suppl_2.ii17
37. Liu PT, Schenk M, Walker VP, Dempsey PW, Kanchanapoomi M,
Wheelwright M, et al. Convergence of IL-1beta and VDR activation pathways
in human TLR2/1-induced antimicrobial responses. PLoS ONE. (2009)
4:e5810. doi: 10.1371/journal.pone.0005810
38. Koeffler HP, Reichel H, Bishop JE, Norman AW. gamma-Interferon
stimulates production of 1,25-dihydroxyvitamin D3 by normal human
macrophages. Biochem Biophys Res Commun. (1985) 127:596–603.
doi: 10.1016/S0006-291X(85)80202-3
39. Bhalla AK, Amento EP, Clemens TL, Holick MF, Krane SM. Specific
high-affinity receptors for 1,25-dihydroxyvitamin D3 in human peripheral
blood mononuclear cells: presence in monocytes and induction in T
lymphocytes following activation. J Clin Endocrinol Metab. (1983) 57:1308–
10. doi: 10.1210/jcem-57-6-1308
40. Provvedini DM, Tsoukas CD, Deftos LJ, Manolagas SC. 1,25-
dihydroxyvitamin D3 receptors in human leukocytes. Science. (1983)
221:1181–3. doi: 10.1126/science.6310748
41. Jeffery LE, Burke F, Mura M, Zheng Y, Qureshi OS, Hewison M, et al.
1,25-Dihydroxyvitamin D3 and IL-2 combine to inhibit T cell production
of inflammatory cytokines and promote development of regulatory T
cells expressing CTLA-4 and FoxP3. J Immunol. (2009) 183:5458–67.
doi: 10.4049/jimmunol.0803217
42. Kim DH, Sabour S, Sagar UN, Adams S, Whellan DJ. Prevalence of
hypovitaminosis D in cardiovascular diseases (from the National Health
and Nutrition Examination Survey 2001 to 2004). Am J Cardiol. (2008)
102:1540–4. doi: 10.1016/j.amjcard.2008.06.067
43. Kendrick J, Targher G, Smits G, Chonchol M. 25-Hydroxyvitamin D
deficiency is independently associated with cardiovascular disease in the
Third National Health and Nutrition Examination Survey. Atherosclerosis.
(2009) 205:255–60. doi: 10.1016/j.atherosclerosis.2008.10.033
44. Manson JE, Cook NR, Lee IM, Christen W, Bassuk SS, Mora S, et al. Vitamin
D supplements and prevention of cancer and cardiovascular disease. N Engl
J Med. (2019) 380:33–44. doi: 10.1056/NEJMoa1809944
Frontiers in Endocrinology | www.frontiersin.org 11 October 2019 | Volume 10 | Article 718
59
Chun et al. Free 25D
45. Ross AC, Manson JE, Abrams SA, Aloia JF, Brannon PM, Clinton SK, 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) 96:53–8. doi: 10.1210/jc.2010-2704
46. Holick MF, Binkley NC, Bischoff-Ferrari HA, Gordon CM, Hanley DA,
Heaney RP, et al. Evaluation, treatment, and prevention of vitamin D
deficiency: an Endocrine Society clinical practice guideline. J Clin Endocrinol
Metab. (2011) 96:1911–30. doi: 10.1210/jc.2011-0385
47. VitaminDCouncil I Tested My Vitamin D Level. What do my results mean?
Available online at: https://www.vitamindcouncil.org/i-tested-my-vitamin-
d-level- what-do- my-results-mean/#.XDeU7i2ZNBw (accessed January 10,
2019).
48. SACN. SACN Vitamin D and Health Report (2016). Available online at:
https://www.gov.uk/government/publications/sacn-vitamin- d-and- health-
report (accessed January 10, 2019).
49. Spiro A, Buttriss JL. Vitamin D: an overview of vitamin D status and intake
in Europe. Nutr Bull. (2014) 39:322–50. doi: 10.1111/nbu.12108
50. Herrmann M, Farrell CL, Pusceddu I, Fabregat-Cabello N, Cavalier E.
Assessment of vitamin D status–a changing landscape. Clin Chem Lab Med.
(2017) 55:3–26. doi: 10.1515/cclm-2016-0264
51. Carlberg C, Haq A. The concept of the personal vitamin D response index. J
Steroid Biochem Mol Biol. (2018) 175:12–7. doi: 10.1016/j.jsbmb.2016.12.011
52. Jones G, Kaufmann M. Vitamin D metabolite profiling using liquid
chromatography-tandem mass spectrometry (LC-MS/MS). J Steroid Biochem
Mol Biol. (2016) 164:110–4. doi: 10.1016/j.jsbmb.2015.09.026
53. Sempos CT, Heijboer AC, Bikle DD, Bollerslev J, Bouillon R, Brannon PM,
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) 84:2194–207. doi: 10.1111/bcp.13652
54. Chapuy MC, Preziosi P, Maamer M, Arnaud S, Galan P, Hercberg S,
et al. Prevalence of vitamin D insufficiency in an adult normal population.
Osteoporos Int. (1997) 7:439–43. doi: 10.1007/s001980050030
55. Holick MF, Siris ES, Binkley N, Beard MK, Khan A, Katzer JT, et al.
Prevalence of vitamin D inadequacy among postmenopausal North
American women receiving osteoporosis therapy. J Clin Endocrinol Metab.
(2005) 90:3215–24. doi: 10.1210/jc.2004-2364
56. Fleet JC. The role of vitamin D in the endocrinology controlling
calcium homeostasis. Mol Cell Endocrinol. (2017) 453:36–45.
doi: 10.1016/j.mce.2017.04.008
57. Tebben PJ, Singh RJ, Kumar R. Vitamin D-mediated hypercalcemia:
mechanisms, diagnosis, and treatment. Endocr Rev. (2016) 37:521–47.
doi: 10.1210/er.2016-1070
58. Kream BE, Jose MJ, DeLuca HF. The chick intestinal cytosol binding protein
for 1,25-dihydroxyvitamin D3: a study of analog binding. Arch Biochem
Biophys. (1977) 179:462–8. doi: 10.1016/0003-9861(77)90134-5
59. Saramaki A, Diermeier S, Kellner R, Laitinen H, Vaisanen S, Carlberg
C. Cyclical chromatin looping and transcription factor association
on the regulatory regions of the p21 (CDKN1A) gene in response
to 1alpha,25-dihydroxyvitamin D3. J Biol Chem. (2009) 284:8073–82.
doi: 10.1074/jbc.M808090200
60. Pike JW. Genome-wide principles of gene regulation by the vitamin D
receptor and its activating ligand. Mol Cell Endocrinol. (2011) 347:3–10.
doi: 10.1016/j.mce.2011.05.012
61. Creo AL, Thacher TD, Pettifor JM, Strand MA, Fischer PR. Nutritional
rickets around the world: an update. Paediatr Int Child Health. (2017)
37:84–98. doi: 10.1080/20469047.2016.1248170
62. Pettifor JM, Prentice A. The role of vitamin D in paediatric bone
health. Best Pract Res Clin Endocrinol Metab. (2011) 25:573–84.
doi: 10.1016/j.beem.2011.06.010
63. Malloy PJ, Feldman D. Genetic disorders and defects in vitamin d action.
Endocrinol Metab Clin North Am. (2010) 39:333–46, table of contents.
doi: 10.1016/j.ecl.2010.02.004
64. Munns CF, Shaw N, Kiely M, Specker BL, Thacher TD, Ozono K,
et al. Global consensus recommendations on prevention and management
of nutritional rickets. J Clin Endocrinol Metab. (2016) 101:394–415.
doi: 10.1210/jc.2015-2175
65. Silverberg SJ, Shane E, Dempster DW, Bilezikian JP. The effects of
vitamin D insufficiency in patients with primary hyperparathyroidism.
Am J Med. (1999) 107:561–7. doi: 10.1016/S0002-9343(99)00
294-6
66. Boudou P, Ibrahim F, Cormier C, Sarfati E, Souberbielle JC. A very high
incidence of low 25 hydroxy-vitamin D serum concentration in a French
population of patients with primary hyperparathyroidism. J Endocrinol
Invest. (2006) 29:511–5. doi: 10.1007/BF03344140
67. Priemel M, von Domarus C, Klatte TO, Kessler S, Schlie J, Meier S, et al.
Bone mineralization defects and vitamin D deficiency: histomorphometric
analysis of iliac crest bone biopsies and circulating 25-hydroxyvitamin D in
675 patients. J Bone Miner Res. (2010) 25:305–12. doi: 10.1359/jbmr.090728
68. Wagner D, Hanwell HE, Vieth R. An evaluation of automated methods
for measurement of serum 25-hydroxyvitamin D. Clin Biochem. (2009)
42:1549–56. doi: 10.1016/j.clinbiochem.2009.07.013
69. Adams JS, Chen H, Chun RF,Nguyen L, Wu S, Ren SY, et al. Novel regulators
of vitamin D action and metabolism: lessons learned at the Los Angeles zoo.
J Cell Biochem. (2003) 88:308–14. doi: 10.1002/jcb.10333
70. Adams JS, Chen H, Chun R, Gacad MA, Encinas C, Ren S, et al. Response
element binding proteins and intracellular vitamin D binding proteins: novel
regulators of vitamin D trafficking, action and metabolism. J Steroid Biochem
Mol Biol. (2004) 89–90:461–5. doi: 10.1016/j.jsbmb.2004.03.016
71. Shinki T, Shiina Y, Takahashi N, Tanioka Y, Koizumi H, Suda T. Extremely
high circulating levels of 1 alpha,25-dihydroxyvitamin D3 in the marmoset,
a new world monkey. Biochem Biophys Res Commun. (1983) 114:452–7.
doi: 10.1016/0006-291X(83)90801-X
72. Adams JS, Gacad MA, Baker AJ, Kheun G, Rude RK. Diminished
internalization and action of 1,25-dihydroxyvitamin D3 in dermal fibroblasts
cultured from New World primates. Endocrinology. (1985) 116:2523–7.
doi: 10.1210/endo-116-6-2523
73. Adams JS, Gacad MA. Phenotypic diversity of the cellular 1,25-
dihydroxyvitamin D3-receptor interaction among different genera
of New World primates. J Clin Endocrinol Metab. (1988) 66:224–9.
doi: 10.1210/jcem-66-1-224
74. Chun RF, Chen H, Boldrick L, Sweet C, Adams JS. Cloning, sequencing,
and functional characterization of the vitamin D receptor in vitamin
D-resistant New World primates. Am J Primatol. (2001) 54:107–18.
doi: 10.1002/ajp.1016
75. Gacad MA, Adams JS. Endogenous blockade of 1,25-dihydroxyvitamin D-
receptor binding in New World primate cells. J Clin Invest. (1991) 87:996–
1001. doi: 10.1172/JCI115108
76. Gacad MA, Adams JS. Specificity of steroid binding in New World primate
B95-8 cells with a vitamin D-resistant phenotype. Endocrinology. (1992)
131:2581–7. doi: 10.1210/endo.131.6.1446602
77. Gacad MA, Chen H, Arbelle JE, LeBon T, Adams JS. Functional
characterization and purification of an intracellular vitamin D-binding
protein in vitamin D-resistant new world primate cells. Amino acid
sequence homology with proteins in the hsp-70 family. J Biol Chem. (1997)
272:8433–40. doi: 10.1074/jbc.272.13.8433
78. Gacad MA, Adams JS. Proteins in the heat shock-70 family specifically bind
25-hydroxyvitamin D3 and 17beta-estradiol. J Clin Endocrinol Metab. (1998)
83:1264–7. doi: 10.1210/jc.83.4.1264
79. Wu S, Chun R, Gacad MA, Ren S, Chen H, Adams JS. Regulation of 1,25-
dihydroxyvitamin d synthesis by intracellular vitamin d binding protein-1.
Endocrinology. (2002) 143:4135. doi: 10.1210/en.2002-220568
80. Wu S, Ren S, Chen H, Chun RF, Gacad MA, Adams JS. Intracellular vitamin
D binding proteins: novel facilitators of vitamin D-directed transactivation.
Mol Endocrinol. (2000) 14:1387–97. doi: 10.1210/mend.14.9.0523
81. Chun R, Gacad MA, Hewison M, Adams JS. Adenosine 5-triphosphate-
dependent vitamin D sterol binding to heat shock protein-70 chaperones.
Endocrinology. (2005) 146:5540–4. doi: 10.1210/en.2005-0579
82. Chun RF, Gacad M, Nguyen L, Hewison M, Adams JS. Co-chaperone
potentiation of vitamin D receptor-mediated transactivation: a role
for Bcl2-associated athanogene-1 as an intracellular-binding protein
for 1,25-dihydroxyvitamin D3. J Mol Endocrinol. (2007) 39:81–9.
doi: 10.1677/JME-07-0042
83. Chun RF, Peercy BE, Adams JS, Hewison M. Vitamin D binding protein
and monocyte response to 25-hydroxyvitamin D and 1,25-dihydroxyvitamin
D: analysis by mathematical modeling. PLoS ONE. (2012) 7:e30773.
doi: 10.1371/journal.pone.0030773
Frontiers in Endocrinology | www.frontiersin.org 12 October 2019 | Volume 10 | Article 718
60
Chun et al. Free 25D
84. Vermeulen A, Verdonck L, Kaufman JM. A critical evaluation of simple
methods for the estimation of free testosterone in serum. J Clin Endocrinol
Metab. (1999) 84:3666–72. doi: 10.1210/jcem.84.10.6079
85. Wilke TJ. A challenge of several concepts of free thyroxin index for assessing
thyroid status in patients with altered thyroid-binding protein capacity. Clin
Chem. (1983) 29:56–9.
86. Rosner W, Auchus RJ, Azziz R, Sluss PM, Raff H. Position statement:
Utility, limitations, and pitfalls in measuring testosterone: an Endocrine
Society position statement. J Clin Endocrinol Metab. (2007) 92:405–13.
doi: 10.1210/jc.2006-1864
87. van Deventer HE, Soldin SJ. The expanding role of tandem mass
spectrometry in optimizing diagnosis and treatment of thyroid disease. Adv
Clin Chem. (2013) 61:127–52. doi: 10.1016/B978-0-12-407680-8.00005-1
88. Nielson CM, Jones KS, Chun RF, Jacobs JM, Wang Y, Hewison M, et al.
Free 25-hydroxyvitamin D: impact of vitamin D binding protein assays on
racial-genotypic associations. J Clin Endocrinol Metab. (2016) 101:2226–34.
doi: 10.1210/jc.2016-1104
89. Bikle DD, Gee E, Halloran B, Haddad JG. Free 1,25-dihydroxyvitamin D
levels in serum from normal subjects, pregnant subjects, and subjects with
liver disease. J Clin Invest. (1984) 74:1966–71. doi: 10.1172/JCI111617
90. Diasource Free 25OH Vitamin D ELISA. Available online at: https://
www.diasource-diagnostics.com/RUO-Products/ImmunoAssays/Bone-
Metabolism/Vitamin-D/Free-25OH- Vitamin-D- ELISA-96-tests (accessed
March 15, 2019).
91. Berg AH, Bhan I, Powe C, Karumanchi SA, Xu D, Thadhani RI. Acute
homeostatic changes following vitamin D2 supplementation. J Endocr Soc.
(2017) 1:1135–49. doi: 10.1210/js.2017-00244
92. Chun RF. New perspectives on the vitamin D binding protein. Cell Biochem
Funct. (2012) 30:445–56. doi: 10.1002/cbf.2835
93. Delanghe JR, Speeckaert R, Speeckaert MM. Behind the scenes of vitamin D
binding protein: more than vitamin D binding. Best Pract Res Clin Endocrinol
Metab. (2015) 29:773–86. doi: 10.1016/j.beem.2015.06.006
94. Rehder DS, Nelson RW, Borges CR. Glycosylation status of vitamin
D binding protein in cancer patients. Protein Sci. (2009) 18:2036–42.
doi: 10.1002/pro.214
95. Nagasawa H, Uto Y, Sasaki H, Okamura N, Murakami A, Kubo S, et al. Gc
protein (vitamin D-binding protein): Gc genotyping and GcMAF precursor
activity. Anticancer Res. (2005) 25:3689–95.
96. Ravnsborg T, Olsen DT, Thysen AH, Christiansen M, Houen G,
Hojrup P. The glycosylation and characterization of the candidate Gc
macrophage activating factor. Biochim Biophys Acta. (2010) 1804:909–17.
doi: 10.1016/j.bbapap.2009.12.022
97. Hollis BW. Comparison of equilibrium and disequilibrium assay conditions
for ergocalciferol, cholecalciferol and their major metabolites. J Steroid
Biochem. (1984) 21:81–6. doi: 10.1016/0022-4731(84)90063-3
98. Van Baelen H, Bouillon R, De Moor P. Vitamin D-binding protein (Gc-
globulin) binds actin. J Biol Chem. (1980) 255:2270–2.
99. Lee WM, Emerson DL, Werner PA, Arnaud P, Goldschmidt-Clermont P,
Galbraith RM. Decreased serum group-specific component protein levels
and complexes with actin in fulminant hepatic necrosis. Hepatology. (1985)
5:271–5. doi: 10.1002/hep.1840050220
100. Swamy N, Ray R. Fatty acid-binding site environments of serum vitamin D-
binding protein and albumin are different. Bioorg Chem. (2008) 36:165–8.
doi: 10.1016/j.bioorg.2008.02.002
101. Kew RR, Webster RO. Gc-globulin (vitamin D-binding protein) enhances
the neutrophil chemotactic activity of C5a and C5a des Arg. J Clin Invest.
(1988) 82:364–9. doi: 10.1172/JCI113596
102. Svasti J, Kurosky A, Bennett A, Bowman BH. Molecular basis for the three
major forms of human serum vitamin D binding protein (group-specific
component). Biochemistry. (1979) 18:1611–7. doi: 10.1021/bi00575a036
103. Constans J, Cleve H. Group-specific component. Report on the First
International Workshop, College de France—July 27–28, 1978–Paris. Hum
Genet. (1979) 48:143–9. doi: 10.1007/BF00286897
104. Kamboh MI, Ferrell RE. Ethnic variation in vitamin D-binding protein (GC):
a review of isoelectric focusing studies in human populations. Hum Genet.
(1986) 72:281–93. doi: 10.1007/BF00290950
105. NCBI dbSNP Short Genetic Variations. Available online at: https://www.ncbi.
nlm.nih.gov/snp/?term=GC (accessed January 10, 2019).
106. Arnaud J, Constans J. Affinity differences for vitamin D metabolites
associated with the genetic isoforms of the human serum carrier protein
(DBP). Hum Genet. (1993) 92:183–8. doi: 10.1007/BF00219689
107. Kawakami M, Imawari M, Goodman DS. Quantitative studies of the
interaction of cholecalciferol (vitamin D3) and its metabolites with different
genetic variants of the serum binding protein for these sterols. Biochem J.
(1979) 179:413–23. doi: 10.1042/bj1790413
108. Bouillon R, van Baelen H, de Moor P. Comparative study of the affinity of
the serum vitamin D-binding protein. J Steroid Biochem. (1980) 13:1029–34.
doi: 10.1016/0022-4731(80)90133-8
109. Boutin B, Galbraith RM, Arnaud P. Comparative affinity of the major
genetic variants of human group-specific component (vitamin D-binding
protein) for 25-(OH) vitamin D. J Steroid Biochem. (1989) 32:59–63.
doi: 10.1016/0022-4731(89)90014-9
110. Chun RF, Lauridsen AL, Suon L, Zella LA, Pike JW, Modlin RL, et al.
Vitamin D-binding protein directs monocyte responses to 25-hydroxy-
and 1,25-dihydroxyvitamin D. J Clin Endocrinol Metab. (2010) 95:3368–76.
doi: 10.1210/jc.2010-0195
111. Jeffery LE, Wood AM, Qureshi OS, Hou TZ, Gardner D, Briggs Z, et al.
Availability of 25-hydroxyvitamin D(3) to APCs controls the balance
between regulatory and inflammatory T cell responses. J Immunol. (2012)
189:5155–64. doi: 10.4049/jimmunol.1200786
112. Kongsbak M, von Essen MR, Levring TB, Schjerling P, Woetmann A, Odum
N, et al. Vitamin D-binding protein controls T cell responses to vitamin D.
BMC Immunol. (2014) 15:35. doi: 10.1186/s12865-014-0035-2
113. Schwartz JB, Gallagher JC, Jorde R, Berg V, Walsh J, Eastell 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) 103:3278–88. doi: 10.1210/jc.2018-00295
114. Powe CE, Evans MK, Wenger J, Zonderman AB, Berg AH, Nalls
M, et al. Vitamin D-binding protein and vitamin D status of black
Americans and white Americans. N Engl J Med. (2013) 369:1991–2000.
doi: 10.1056/NEJMoa1306357
115. Denburg MR, Hoofnagle AN, Sayed S, Gupta J, de Boer IH, Appel LJ,
et al. 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) 31:1128–36. doi: 10.1002/jbmr.2829
116. Lauridsen AL, Vestergaard P, Nexo E. Mean serum concentration of vitamin
D-binding protein (Gc globulin) is related to the Gc phenotype in women.
Clin Chem. (2001) 47:753–6.
117. Schwartz JB, Lai J, Lizaola B, Kane L, Markova S, Weyland P, et al.
A comparison of measured and calculated free 25(OH) vitamin D
levels in clinical populations. J Clin Endocrinol Metab. (2014) 99:1631–7.
doi: 10.1210/jc.2013-3874
118. Henderson CM, Fink SL, Bassyouni H, Argiropoulos B, Brown L, Laha TJ,
et al. Vitamin D-binding protein deficiency and homozygous deletion of the
GC gene. N Engl J Med. (2019) 380:1150–7. doi: 10.1056/NEJMoa1807841
119. Safadi FF, Thornton P, Magiera H, Hollis BW, Gentile M, Haddad JG, et al.
Osteopathy and resistance to vitamin D toxicity in mice null for vitamin D
binding protein. J Clin Invest. (1999) 103:239–51. doi: 10.1172/JCI5244
120. 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) 149:3656–67. doi: 10.1210/en.2008-0042
121. Chun RF, Hernandez I, Pereira R, Swinkles L, Huijs T, Zhou R, et al.
Differential responses to vitamin D2 and vitamin D3 are associated with
variations in free 25-hydroxyvitamin D. Endocrinology. (2016) 157:3420–30.
doi: 10.1210/en.2016-1139
122. Southworth LO, Holick MF, Chen TC, Kunz TH. Effects of sunlight on
behavior and 25-hydroxyvitamin D levels in two species of Old World fruit
bats. Dermatoendocrinology. (2013) 5:192–8. doi: 10.4161/derm.24020
123. Vieth R, Kessler MJ, Pritzker KP. Species differences in the binding kinetics
of 25-hydroxyvitamin D3 to vitamin D binding protein. Can J Physiol
Pharmacol. (1990) 68:1368–71. doi: 10.1139/y90-207
124. Adams JS, Hewison M. Unexpected actions of vitamin D: new perspectives
on the regulation of innate and adaptive immunity. Nat Clin Pract Endocrinol
Metab. (2008) 4:80–90. doi: 10.1038/ncpendmet0716
Frontiers in Endocrinology | www.frontiersin.org 13 October 2019 | Volume 10 | Article 718
61
Chun et al. Free 25D
125. Yu C, Xue H, Wang L, Chen Q, Chen X, Zhang Y, et al. Serum bioavailable
and free 25-hydroxyvitamin D levels, but not its total level, are associated
with the risk of mortality in patients with coronary artery disease. Circ Res.
(2018) 123:996–1007. doi: 10.1161/CIRCRESAHA.118.313558
126. Strisciuglio C, Cenni S, Giugliano FP, Miele E, Cirillo G, Martinelli M,
et al. The role of inflammation on vitamin D levels in a cohort of pediatric
patients with inflammatory bowel disease. J Pediatr Gastroenterol Nutr.
(2018) 67:501–6. doi: 10.1097/MPG.0000000000002049
127. Sauer CG, Loop MS, Venkateswaran S, Tangpricha V, Ziegler TR, Dhawan
A, et al. Free and bioavailable 25-hydroxyvitamin D concentrations are
associated with disease activity in pediatric patients with newly diagnosed
treatment naive ulcerative colitis. Inflamm Bowel Dis. (2018) 24:641–50.
doi: 10.1093/ibd/izx052
128. Lee CC, Young KA, Norris JM, Rotter JI, Liu Y, Lorenzo C, et al.
Association of directly measured plasma free 25(OH)D with insulin
sensitivity and secretion: the IRAS Family Study. J Clin Endocrinol Metab.
(2017) 102:2781–8. doi: 10.1210/jc.2017-00039
129. Kane L, Moore K, Lutjohann D, Bikle D, Schwartz JB. Vitamin D3 effects
on lipids differ in statin and non-statin-treated humans: superiority of free
25-OH D levels in detecting relationships. J Clin Endocrinol Metab. (2013)
98:4400–9. doi: 10.1210/jc.2013-1922
130. Altinova AE, Ozkan C, Akturk M, Gulbahar O, Yalcin M, Cakir N,
et al. Vitamin D-binding protein and free vitamin D concentrations in
acromegaly. Endocrine. (2016) 52:374–9. doi: 10.1007/s12020-015-0789-1
131. Lima JJ, Castro M, King TS, Lang JE, Ortega VE, Peters SP, et al. Association
of free vitamin D3 concentrations and asthma treatment failures in the
VIDA Trial. Ann Allergy Asthma Immunol. (2018) 121:444–450 e441.
doi: 10.1016/j.anai.2018.06.001
132. Andersen SW, Shu XO, Cai Q, Khankari NK, Steinwandel MD, Jurutka
PW, et al. Total and free circulating vitamin D and vitamin D-binding
protein in relation to colorectal cancer risk in a prospective study of
African Americans. Cancer Epidemiol Biomarkers Prev. (2017) 26:1242–7.
doi: 10.1158/1055-9965.EPI-17-0133
133. Powe CE, Ricciardi C, Berg AH, Erdenesanaa D, Collerone G, Ankers E, et al.
Vitamin D-binding protein modifies the vitamin D-bone mineral density
relationship. J Bone Miner Res. (2011) 26:1609–16. doi: 10.1002/jbmr.387
134. Bhan I, Powe CE, Berg AH, Ankers E, Wenger JB, Karumanchi SA, et al.
Bioavailable vitamin D is more tightly linked to mineral metabolism than
total vitamin D in incident hemodialysis patients. Kidney Int. (2012) 82:84–9.
doi: 10.1038/ki.2012.19
135. R&DBiosystems Improved Sensitivity of Vitamin D BP ELISA Kits. Available
online at: https://www.rndsystems.com/product-highlight/improved-
sensitivity-vitamin- d-bp-elisa-kits (accessed March 27, 2019).
136. Aloia J, Dhaliwal R, Mikhail M, Shieh A, Stolberg A, Ragolia L, et al. Free
25(OH)D and calcium absorption, PTH, and markers of bone turnover. J
Clin Endocrinol Metab. (2015) 100:4140–5. doi: 10.1210/jc.2015-2548
137. Peris P, Filella X, Monegal A, Guanabens N, Foj L, Bonet M, et al.
Comparison of total, free and bioavailable 25-OH vitamin D determinations
to evaluate its biological activity in healthy adults: the LabOscat study.
Osteoporos Int. (2017) 28:2457–64. doi: 10.1007/s00198-017-4062-8
138. Walsh JS, Evans AL, Bowles S, Naylor KE, Jones KS, Schoenmakers I,
et al. Free 25-hydroxyvitamin D is low in obesity, but there are no
adverse associations with bone health. Am J Clin Nutr. (2016) 103:1465–71.
doi: 10.3945/ajcn.115.120139
139. Lai JC, Bikle DD, Lizaola B, Hayssen H, Terrault NA, Schwartz JB. Total
25(OH) vitamin D, free 25(OH) vitamin D and markers of bone turnover
in cirrhotics with and without synthetic dysfunction. Liver Int. (2015)
35:2294–300. doi: 10.1111/liv.12819
140. Lopez-Molina M, Santillan C, Murillo M, Valls A, Bosch L, Bel
J, et al. Measured free 25-hydroxyvitamin D in healthy children
and relationship to total 25-hydroxyvitamin D, calculated free 25-
hydroxyvitamin D and vitamin D binding protein. Clin Biochem. (2018)
61:23–7. doi: 10.1016/j.clinbiochem.2018.08.007
141. Alzaman NS, Dawson-Hughes B, Nelson J, D’Alessio D, Pittas AG. Vitamin
D status of black and white Americans and changes in vitamin D metabolites
after varied doses of vitamin D supplementation. Am J Clin Nutr. (2016)
104:205–14. doi: 10.3945/ajcn.115.129478
142. Sollid ST, Hutchinson MY, Berg V, Fuskevag OM, Figenschau Y, Thorsby
PM, et al. Effects of vitamin D binding protein phenotypes and vitamin
D supplementation on serum total 25(OH)D and directly measured free
25(OH)D. Eur J Endocrinol. (2016) 174:445–52. doi: 10.1530/EJE-15-1089
143. Tsuprykov O, Buse C, Skoblo R, Haq A, Hocher B. Reference intervals for
measured and calculated free 25-hydroxyvitamin D in normal pregnancy. J
Steroid Biochem Mol Biol. (2018) 181:80–7. doi: 10.1016/j.jsbmb.2018.03.005
144. Schwartz JB, Kane L, Bikle D. Response of vitamin D concentration
to vitamin D3 administration in older adults without sun exposure:
a randomized double-blind trial. J Am Geriatr Soc. (2016) 64:65–72.
doi: 10.1111/jgs.13774
145. Shieh A, Chun RF, Ma C, Witzel S, Meyer B, Rafison B, et al. Effects of
high-dose vitamin D2 versus D3 on total and free 25-hydroxyvitamin D and
markers of calcium balance. J Clin Endocrinol Metab. (2016) 101:3070–8.
doi: 10.1210/jc.2016-1871
146. Shieh A, Ma C, Chun RF, Wittwer-Schegg J, Swinkels L, Huijs T, et al.
Associations between change in total and free 25-hydroxyvitamin D with
24,25-dihydroxyvitamin D and parathyroid hormone. J Clin Endocrinol
Metab. (2018) 103:3368–75. doi: 10.1210/jc.2018-00515
147. Wang X, Shapses SA, Al-Hraishawi H. Free and bioavailable 25-
hydroxyvitamin D levels in patients with primary hyperparathyroidism.
Endocr Pract. (2017) 23:66–71. doi: 10.4158/EP161434.OR
148. Szabo B, Tabak AG, Toldy E, Szekeres L, Szili B, Bakos B, et al. The role
of serum total and free 25-hydroxyvitamin D and PTH values in defining
vitamin D status at the end of winter: a representative survey. J Bone Miner
Metab. (2017) 35:83–90. doi: 10.1007/s00774-015-0729-4
149. Gopal-Kothandapani JS, Evans LF, Walsh JS, Gossiel F, Rigby AS, Eastell
R, et al. Effect of vitamin D supplementation on free and total vitamin
D: a comparison of Asians and Caucasians. Clin Endocrinol (Oxf). (2019)
90:222–31. doi: 10.1111/cen.13825
150. Best CM, Pressman EK, Queenan RA, Cooper E, O’Brien KO. Longitudinal
changes in serum vitamin D binding protein and free 25-hydroxyvitamin D
in a multiracial cohort of pregnant adolescents. J Steroid Biochem Mol Biol.
(2018) 186:79–88. doi: 10.1016/j.jsbmb.2018.09.019
151. Jemielita TO, Leonard MB, Baker J, Sayed S, Zemel BS, Shults J, et al.
Association of 25-hydroxyvitamin D with areal and volumetric measures
of bone mineral density and parathyroid hormone: impact of vitamin
D-binding protein and its assays. Osteoporos Int. (2016) 27:617–26.
doi: 10.1007/s00198-015-3296-6
152. Alwan A, Rizkallah M, Maalouf G, Matta J, Frenn F, Berro AJ, et al.
Positive correlations between free vitamin D and bone variables in
a group of young Lebanese men. J Clin Densitom. (2018) 21:459–61.
doi: 10.1016/j.jocd.2018.02.001
153. Michaelsson K, Rasmusson A, Wolk A, Byberg L, Mitchell A, Melhus H. The
free hormone hypothesis: is free serum 25-hydroxyvitamin D a better marker
for bone mineral density in older women? JBMR Plus. (2018) 2:367–74.
doi: 10.1002/jbm4.10059
154. Segersten U, Correa P, Hewison M, Hellman P, Dralle H, Carling T,
et al. 25-hydroxyvitamin D(3)-1alpha-hydroxylase expression in normal and
pathological parathyroid glands. J Clin Endocrinol Metab. (2002) 87:2967–72.
doi: 10.1210/jcem.87.6.8604
155. Ritter CS, Armbrecht HJ, Slatopolsky E, Brown AJ. 25-Hydroxyvitamin D(3)
suppresses PTH synthesis and secretion by bovine parathyroid cells. Kidney
Int. (2006) 70:654–9. doi: 10.1038/sj.ki.5000394
156. Ritter CS, Brown AJ. Direct suppression of Pth gene expression by the
vitamin D prohormones doxercalciferol and calcidiol requires the vitamin
D receptor. J Mol Endocrinol. (2011) 46:63–6. doi: 10.1677/JME-10-0128
157. Weaver CM, Gordon CM, Janz KF, Kalkwarf HJ, Lappe JM, Lewis R,
et al. The National Osteoporosis Foundation’s position statement on peak
bone mass development and lifestyle factors: a systematic review and
implementation recommendations. Osteoporos Int. (2016) 27:1281–386.
doi: 10.1007/s00198-015-3440-3
158. Malmstroem S, Rejnmark L, Imboden JB, Shoback DM, Bikle DD. Current
assays to determine free 25-hydroxyvitamin D in serum. J AOAC Int. (2017)
100:1323–7. doi: 10.5740/jaoacint.17-0085
159. Ray JA, Kushnir MM, Bunker A, Rockwood AL, Meikle AW. Direct
measurement of free estradiol in human serum by equilibrium dialysis-liquid
Frontiers in Endocrinology | www.frontiersin.org 14 October 2019 | Volume 10 | Article 718
62
Chun et al. Free 25D
chromatography-tandem mass spectrometry and reference intervals
of free estradiol in women. Clin Chim Acta. (2012) 413:1008–14.
doi: 10.1016/j.cca.2012.02.028
160. Welsh KJ, Soldin SJ. Diagnosis of endocrine disease: how reliable are free
thyroid and total T3 hormone assays? Eur J Endocrinol. (2016) 175:R255–63.
doi: 10.1530/EJE-16-0193
161. Fiers T, Wu F, Moghetti P, Vanderschueren D, Lapauw B, Kaufman JM.
Reassessing free-testosterone calculation by liquid chromatography-
tandem mass spectrometry direct equilibrium dialysis. J Clin
Endocrinol Metab. (2018) 103:2167–74. doi: 10.1210/jc.2017-
02360
Conflict of Interest: The authors declare that the research was conducted in the
absence of any commercial or financial relationships that could be construed as a
potential conflict of interest.
Copyright © 2019 Chun, Shieh, Gottlieb, Yacoubian, Wang, Hewison and Adams.
This is an open-access article distributed under the terms of the Creative Commons
Attribution License (CC BY). The use, distribution or reproduction in other forums
is permitted, provided the original author(s) and the copyright owner(s) are credited
and that the original publication in this journal is cited, in accordance with accepted
academic practice. No use, distribution or reproduction is permitted which does not
comply with these terms.
Frontiers in Endocrinology | www.frontiersin.org 15 October 2019 | Volume 10 | Article 718
63
Advantages
of publishing
in Frontiers
OPEN ACCESS
Articles are free to read
for greatest visibility
and readership
EXTENSIVE PROMOTION
Marketing
and promotion
of impactful research
DIGITAL PUBLISHING
Articles designed
for optimal readership
across devices
LOOP RESEARCH NETWORK
Our network
increases your
article’s readership
Frontiers
Avenue du Tribunal-Fédéral 34
1005 Lausanne | Switzerland
Visit us: www.frontiersin.org
Contact us: info@frontiersin.org | +41 21 510 17 00
FAST PUBLICATION
Around 90 days
from submission
to decision
90
IMPACT METRICS
Advanced article metrics
track visibility across
digital media
FOLLOW US
@frontiersin
TRANSPARENT PEER-REVIEW
Editors and reviewers
acknowledged by name
on published articles
HIGH QUALITY PEER-REVIEW
Rigorous, collaborative,
and constructive
peer-review
REPRODUCIBILITY OF
RESEARCH
Support open data
and methods to enhance
research reproducibility
Article
Background: Higher levels of physical activity are related to higher 25-(OH)D levels. Total 25-(OH)D (25-(OH)DT) are routinely used in clinical practice to assess vitamin D, however novel biomarkers are currently being investigated as free 25-(OH)D (25-(OH)DF) or vitamin D metabolite ratios (VMRs). The primary aim of our study was to assess 25-(OH)DF, vitamin D metabolites and VMRs in inactive men and athletes. A secondary aim was to check whether regular physical activity influence on vitamin D metabolome. A tertiary aim was to determine the relationship between 25-(OH)DT, 25-(OH)DF, vitamin D binding protein (VDBP), vitamin D metabolites and VMRs in this cohort. Methods: A total of 69 participants (27 inactive men, 18 indoor and 24 outdoor athletes) participated in the study. Vitamin D metabolites (25-(OH)DT, 24,25-(OH)2D3, 3-epi-25-(OH)D3, and 1,25-(OH)2D) were assessed using LC-MS/MS. The 25-(OH)DF concentration was calculated based on serum albumin and VDBP levels. Results: There were no differences in vitamin D metabolites and VMRs between inactive men and between the two groups of athletes. We showed a strong relationship between 25-(OH)DT, 25-(OH)DF and 24,25-(OH)D3, 3-epi-25(OH)D3 in each group. Analysis showed that 25-(OH)DT, 25-(OH)DF inversely associated with 25-(OH)D3:24,25-(OH)2D3, 25-(OH)D3:3-epi-25-(OH)D3, 1,25-(OH)2D:24,25-(OH)2D3 ratios in inactive men and athletes (indoor and outdoor). Conclusions: On the basis of our results, we concluded that regular long-term physical activity has no effect on the concentration of vitamin D metabolites at rest. Furthermore, free vitamin D does not correlate more strongly with vitamin D metabolites and VMRs compared to total.
Article
Vitamin D supplements have long been advocated for people with chronic kidney disease (CKD) based on data from observational studies among the general population and also people with CKD. These data consistently suggested that higher circulating concentrations of 25-hydroxyvitamin D are associated with improved fracture, cardiovascular, cancer, and mortality outcomes. In the last few years, large clinical trials have been conducted to assess the effects of vitamin D supplements on a range of clinically relevant outcomes. Most of these studies were performed in the general population but also enrolled people with CKD. Virtually all these trials were negative and contradicted the observational data. In this review, the key observational data and clinical trials are summarized and potential explanations for the discrepancies between these studies are discussed.
Article
Full-text available
Studies in vitro have demonstrated a key molecular role for 1,25-dihydroxyvitamin D (1,25D) in skeletal muscle function, with vitamin D-deficiency (low serum 25-hydroxyvitamin D, 25D) being associated with muscle pain and weakness. Despite this, an understanding of the overall role of vitamin D in muscle health (particularly the impact of vitamin D-related genetic variants) has yet to be fully resolved, relative to more well-studied targets such as the skeleton. Thus, we aimed to review existing studies that have investigated relationships between skeletal muscle function and single nucleotide polymorphisms (SNPs) within vitamin D-related genes. A systematic review of papers published between January 2000 and June 2022 on PubMed, EMBASE and Web of Science pertaining to association between functionally relevant vitamin D receptor genetic variants and variants within genes of the vitamin D pathway and skeletal muscle function/outcomes was performed. 21 articles were included in the review for final analysis, of which 20 only studied genetic variation of the VDR gene. Of the included articles, 81% solely included participants aged ≥50 years and of the 9 studies that did not only include White individuals, only 2 included Black participants. Within the vitamin D system, the VDR gene is the primary gene of which associations between polymorphisms and muscle function have been investigated. VDR polymorphisms have been significantly associated with muscle phenotypes in two or more studies. Of note A1012G was significantly associated with higher handgrip strength, but the results for other SNPs were notably variable between studies. While the lack of definitive evidence and study heterogeneity makes it difficult to draw conclusions, the findings of this review highlight a need for improvements with regards to the use of more diverse study populations, i.e., inclusion of Black individuals and other people of colour, and expanding research scope beyond the VDR gene.
Article
Full-text available
Clinical mastitis (CM) is an inflammatory disease occurring in the mammary glands of lactating cows. CM is under genetic control, and a prominent CM resistance QTL located on chromosome 6 was reported in various dairy cattle breeds. Nevertheless, the biological mechanism underpinning this QTL has been lacking. Herein, we mapped, fine-mapped, and discovered the putative causal variant underlying this CM resistance QTL in the Dutch dairy cattle population. We identified a ~12 kb multi-allelic copy number variant (CNV), that is in perfect linkage disequilibrium with a lead SNP, as a promising candidate variant. By implementing a fine-mapping and through expression QTL mapping, we showed that the group-specific component gene (GC), a gene encoding a vitamin D binding protein, is an excellent candidate causal gene for the QTL. The multiplicated alleles are associated with increased GC expression and low CM resistance. Ample evidence from functional genomics data supports the presence of an enhancer within this CNV, which would exert cis-regulatory effect on GC. We observed that strong positive selection swept the region near the CNV, and haplotypes associated with the multiplicated allele were strongly selected for. Moreover, the multiplicated allele showed pleiotropic effects for increased milk yield and reduced fertility, hinting that a shared underlying biology for these effects may revolve around the vitamin D pathway. These findings together suggest a putative causal variant of a CM resistance QTL, where a cis-regulatory element located within a CNV can alter gene expression and affect multiple economically important traits.
Article
Full-text available
The role of vitamin D in psoriasis remains contradictory despite the fact that vitamin D analogues constitute an established treatment for psoriasis. It has been proposed that the ability of vitamin D to exert anti-inflammatory effects might not depend solely on the concentration of serum 25(OH)D but also on the concentration of vitamin D-binding protein (DBP). High concentrations of DBP might diminish vitamin D´s biologic action. The aims of this study were (i) to analyze the serum levels of DBP, total and calculated free 25(OH)D in patients with psoriasis and compare the results with healthy control and (ii) to study the effect of UVB phototherapy on DBP levels. Caucasian subjects (n = 68) with active plaque psoriasis were compared with a population-based sample of men and women (n = 105), matched for age and sex. Season of enrollment was taken into consideration. The patients were also studied before and after UVB phototherapy. The severity of the disease was calculated as Psoriasis Area Severity Index (PASI). DBP, free 25(OH)D index and total 25(OH)D were higher in patients with psoriasis compared with controls (P = 0.004, P = 0.045 and P < 0.0001, respectively). DBP did not change after phototherapy, whereas 25(OH)D increased and intact parathyroid hormone decreased (p < 0.001 for both). Psoriasis improved and PASI decreased after phototherapy (p < 0.001). There was no correlation between DBP and 25(OH)D or between DBP and PASI. Measurement of DBP is recommended when evaluating vitamin D status in patients with psoriasis. High DBP levels in psoriasis imply a disturbed vitamin D pathway that warrants further investigation. Direct measurement of free 25(OH)D, instead of total 25(OH)D that circumvents abnormally high levels of DBP, could be considered.
Article
Full-text available
Background: Lifestyle therapy with resistance training is a potent measure to counteract age-related loss in muscle strength and mass. Unfortunately, many individuals fail to respond in the expected manner. This phenomenon is particularly common among older adults and those with chronic diseases (e.g. chronic obstructive pulmonary disease, COPD) and may involve en-docrine variables such as vitamin D. At present, the effects of vitamin D supplementation on responses to resistance training remain largely unexplored. Methods: Ninety-five male and female participants (healthy, n = 71; COPD, n = 24; age 68 ± 5 years) were randomly assigned to receive either vitamin D 3 or placebo supplementation for 28 weeks in a double-blinded manner (latitude 61°N, September-May). Seventy-eight participants completed the RCT, which was initiated by 12 weeks of supplementation-only (two weeks with 10 000 IU/day, followed by 2000 IU/day), followed by 13 weeks of combined supplementation (2000 IU/day) and supervised whole-body resistance training (twice weekly), interspersed with testing and measurements. Outcome measures included multiple assessments of muscle strength (n variables = 7), endurance performance (n = 6), and muscle mass (n = 3, legs, primary), as well as muscle quality (legs), muscle biology (m. vastus lateralis; muscle fibre characteristics, transcriptome), and health-related variables (e.g. visceral fat mass and blood lipid profile). For main outcome domains such as muscle strength and muscle mass, weighted combined factors were calculated from the range of singular assessments. Results: Overall, 13 weeks of resistance training increased muscle strength (13% ± 8%), muscle mass (9% ± 8%), and endurance performance (one-legged, 23% ± 15%; whole-body, 8% ± 7%), assessed as weighted combined factors, and were associated with changes in health variables (e.g. visceral fat, À6% ± 21%; [LDL] serum , À4% ± 14%) and muscle tissue characteristics such as fibre type proportions (e.g. IIX, À3% points), myonuclei per fibre (30% ± 65%), total RNA/rRNA abundances (15%/ 6-19%), and transcriptome profiles (e.g. 312 differentially expressed genes). Vitamin D 3 supplementation did not affect training-associated changes for any of the main outcome domains, despite robust increases in [25(OH)D] serum (Δ49% vs. placebo). No conditional effects were observed for COPD vs. healthy or pre-RCT [25(OH)D] serum. In secondary analyses, vitamin D 3 affected expression of gene sets involved in vascular functions in muscle tissue and strength gains in participants with high fat mass, which advocates further study. Conclusions: Vitamin D3 supplementation did not affect muscular responses to resistance training in older adults with or without COPD.
Article
Full-text available
Importance: Deficient (ie, <20 ng/mL) or insufficient (ie, 20 to <30 ng/mL) 25-hydroxyvitamin D (also known as calcifediol) levels are more common in Black individuals than White individuals and are associated with increased coronavirus disease 2019 (COVID-19) risk. Whether COVID-19 risk is associated with differences in vitamin D levels of 30 ng/mL or greater is not known. Objective: To examine whether COVID-19 test results are associated with differences in vitamin D levels of 30 ng/mL or greater, including for White individuals and for Black individuals. Design, setting, and participants: This retrospective cohort study was conducted at an academic medical center in Chicago, Illinois. Participants included individuals with data on vitamin D level within 365 days before COVID-19 testing, which was conducted from March 3 to December 30, 2020. Data were analyzed from September 11, 2020, to February 5, 2021. Exposures: The last vitamin D level before COVID-19 testing was categorized as less than 20 ng/mL (ie, deficient), 20 to less than 30 ng/mL (ie, insufficient), 30 to less than 40 ng/mL, or 40 ng/mL or greater. Treatment was defined by vitamin D type and dose 14 days before COVID-19 testing and treatment changes after last vitamin D level. Main outcomes and measures: The main outcome was a positive result for COVID-19 in polymerase chain reaction testing. Multivariable analyses tested whether previously measured vitamin D level was associated with having test results positive for COVID-19 in White individuals and in Black individuals, controlling for months and treatment changes since the vitamin D level was measured, as well as demographic characteristics and comorbidity indicators. Results: A total of 4638 individuals (mean [SD] age 52.8 [19.5] years; 3205 [69%] women) had data for a vitamin D level within 1 year before COVID-19 testing, including 2288 (49%) Black individuals, 1999 (43%) White individuals, and 351 individuals (8%) who were another race/ethnicity (eg, Asian, Mideast Indian, >1 race). Stratified by vitamin D level, 1251 individuals (27%) had less than 20 ng/mL, 1267 individuals (27%) had 20 to less than 30 ng/mL, 1023 individuals (22%) had 30 to less than 40 ng/mL, and 1097 individuals (24%) had 40 ng/mL or greater. Lower vitamin D levels were more common in Black individuals (<20 ng/mL: 829 of 2288 Black individuals [36%]) than White individuals (<20 ng/mL: 315 of 1999 White individuals [16%]). A total of 333 individuals (7%) had test results positive for COVID-19, including 102 White individuals (5%) and 211 Black individuals (9%). Multivariate analysis controlling for time since last vitamin D level measurement was used to estimate the outcomes associated with levels 14 days before COVID-19 testing. A positive test result for COVID-19 was not significantly associated with vitamin D levels in White individuals but was associated with vitamin D levels in Black individuals (compared with ≥40 ng/mL: <20 ng/mL incidence rate ratio [IRR], 2.55 [95% CI, 1.26-5.15]; P = .009; 20 to <30 ng/mL IRR, 1.69 [95% CI, 0.75-3.84]; P = .21; 30 to <40 ng/mL IRR, 2.64 [95% CI, 1.24-5.66]; P = .01). Stratified by vitamin D level, estimated COVID-19 positivity rates in Black individuals were 9.72% (95% CI, 6.74%-13.41%) for individuals with a vitamin D level less than 20 ng/mL, 6.47% (95% CI, 3.33%-10.28%) for individuals with a vitamin D level of 20 to less than 30 ng/mL, 10.10% (95% CI, 6.00%-15.47%) for individuals with a vitamin D level of 30 to less than 40 ng/mL, and 3.82% (95% CI, 1.78%-6.68%) for individuals with a vitamin D level of 40 ng/mL or higher. Multivariate analysis in individuals with a vitamin D level of 30 ng/mL or greater found that the IRR of a positive COVID-19 test result was 0.97 (95% CI, 0.94-0.99; P = .008) per 1-ng/mL increase in vitamin D overall and 0.95 (95% CI, 0.91-0.98; P = .003) per 1-ng/mL increase in vitamin D in Black individuals. Conclusions and relevance: In this single-center retrospective cohort study, COVID-19 risk increased among Black individuals with vitamin D level less than 40 ng/mL compared with those with 40 ng/mL or greater and decreased with increasing levels among individuals with levels greater than 30 ng/mL. No significant associations were noted for White individuals. Randomized clinical trials should examine whether increasing vitamin D level to greater than 40 ng/mL affects COVID-19 risk.
Article
Full-text available
Vitamin D is important for bone health, but may also have extra-skeletal effects. Vitamin D and its binding protein DBP have immunological effects and may therefore be important in the development of type 1 diabetes (T1DM), and low serum levels of 25-hydroxyvitamin D (25(OH)D) are associated with later development of type 2 diabetes (T2DM). However, it has so far been difficult to convincingly show an effect of vitamin D supplementation on prevention or treatment of diabetes. The serum level of 25(OH)D has traditionally been used as a marker of a subject's vitamin D status. This measurement includes both 25(OH)D bound to DBP and albumin as well as the free from of 25(OH)D. However, according to the free hormone hypothesis, the free form is the biologically active. Previously the free form of 25(OH)D had to be calculated based on measurements of 25(OH)D, DBP, and albumin, but recently a method for direct measurement of free 25(OH)D has become commercially available. This is important in clinical conditions where the amount of DBP is affected, and has caused a renewed interest in which vitamin D metabolite to measure in clinical situations. In the present review the relations between DBP, total and free 25(OH)D in T1DM and T2DM are described.
Article
Full-text available
Neutrophils have always been considered as uncomplicated front-line troopers of the innate immune system equipped with limited proinflammatory duties. Yet recently, the role of the neutrophil has been undergoing a rejuvenation of sorts. Neutrophils are now considered complex cells capable of a significant array of specialized functions, and as an effector of the innate immune response, they are able to regulate many processes such as acute injury and repair, cancer, autoimmunity, and chronic inflammatory processes. Furthermore, evidence exists to indicate that neutrophils also contribute to adaptive immunity by aiding the development of specific adaptive immune responses or guiding the subsequent adaptive immune response. With this revived interest in neutrophils and their many novel functions, it is prudent to review what is currently known about neutrophils and, even more importantly, understand what information is lacking. We discuss the essential features of the neutrophil, from its origins, lifespan, subsets, margination and sequestration of the neutrophil to the death of the neutrophil. We highlight neutrophil recruitment to both infected and injured tissues and outline differences in recruitment of neutrophils between different tissues. Finally, we examine how neutrophils use different mechanisms to either bolster protective immune responses or negatively cause pathological outcomes at different locations.
Article
Full-text available
Background: Randomised controlled trials of adjunctive vitamin D in pulmonary tuberculosis (TB) treatment have yielded conflicting results. Individual participant data meta-analysis could identify factors explaining this variation. Methods: We meta-analysed individual participant data from randomised controlled trials of vitamin D in patients receiving antimicrobial therapy for pulmonary TB. Primary outcome was time to sputum culture conversion. Secondary outcomes were time to sputum smear conversion, mean 8-week weight and incidence of adverse events. Pre-specified subgroup analyses were done according to baseline vitamin D status, age, sex, drug susceptibility, HIV status, extent of disease and vitamin D receptor genotype. Results: Individual participant data were obtained for 1850 participants in eight studies. Vitamin D did not influence time to sputum culture conversion overall (adjusted HR 1.06, 95% CI 0.91-1.23), but it did accelerate sputum culture conversion in participants with multidrug-resistant pulmonary TB (adjusted HR 13.44, 95% CI 2.96-60.90); no such effect was seen in those whose isolate was sensitive to rifampicin and/or isoniazid (adjusted HR 1.02, 95% CI 0.88-1.19; p-value for interaction=0.02). Vitamin D accelerated sputum smear conversion overall (adjusted HR 1.15, 95% CI 1.01-1.31), but did not influence other secondary outcomes. Conclusions: Vitamin D did not influence time to sputum culture conversion overall, but it accelerated sputum culture conversion in patients with multidrug-resistant pulmonary TB.
Article
Full-text available
Purpose: We aimed to investigate the status of serum 25-hydroxyvitamin D [25(OH)D] among Chinese postmenopausal women in a multicenter cross-sectional study. Methods: Non-institutionalized postmenopausal women aged ≥55 years were recruited from urban and rural areas in 7 geographically different regions in China. Subject enrollment was executed during the summer and the winter. Vitamin D insufficiency and deficiency were defined as 25(OH)D < 30 and< 20 ng/ml, and was measured by liquid chromatography-tandem mass spectrometry. Women were referred to a dual-energy x-ray absorptiometry (DXA) if they had a medium-to-high fracture risk suggested by Osteoporosis Self-Assessment Tool for Asians (OSTA). Results: Among all subjects, 91.2% (1,535/1,684, 95%CI: 89.7, 92.5) had vitamin D insufficiency and 61.3% had vitamin D deficiency (1,033/1,684, 95%CI: 59.0, 63.7). The prevalence of vitamin D deficiency was significantly higher in urban dwellers (64.9 vs. 57.7% in rural, P = 0.002) and in winter-enrolled subjects (84.7 vs. 41.3% in summer, P < 0.0001). The prevalence of vitamin D inadequacy did not increase in trend by latitude and was numerically lower in women who had high fracture risk and osteoporosis. A non-curvilinear change of intact parathyroid hormone (iPTH) levels was observed at 25(OH)D >16.78 ng/mL. Conclusions: The prevalence of vitamin D inadequacy was remarkable among Chinese postmenopausal women and independent of fracture risk assessed by OSTA or osteoporosis suggested by DXA. Winter season, urban residence, however not latitude, were significantly associated with a higher likelihood of vitamin D deficiency. Optimal vitamin D status for iPTH and bone-related outcomes merits further investigation in this population.
Article
Actin is an abundant intracellular protein that is released into the blood during tissue injury and its injection into rats causes microthrombi to form in the vasculature. This report and others have shown that actin filaments are able to aggregate platelets in an adenosine diphosphate (ADP)-dependent manner. The effects on this process of two plasma actin-binding proteins, vitamin D-binding protein (DBP) and gelsolin, were examined separately and together. The addition of DBP, a monomer-binding protein, to actin filaments did not affect their ability to induce platelet aggregation. However, severing of actin filaments with gelsolin resulted in an increased degree of platelet aggregation. Preincubation of F-actin with both gelsolin and DBP resulted in a significant inhibition of aggregation. The effects of DBP and gelsolin on actin-induced aggregation paralleled their effects on exchange of actin-bound adenine nucleotides. DBP inhibited 1, N6- ethenoadenosine 5′ triphosphate (epsilon-ATP) exchange with G-actin but not with F-actin. Gelsolin increased epsilon-ATP exchange with F-actin, which was largely abrogated by the addition of DBP. These results suggest that gelsolin's severing (and subsequent capping) of actin filaments not only results in an increase in the number of pointed filament ends but also in the dissociation of actin monomers containing ADP. Phalloidin, which stabilizes actin filaments while decreasing both monomer and nucleotide exchange, inhibited actin-induced aggregation, as well, indicating that depolymerization of actin filaments is not required to inhibit aggregation. Platelet activation by either G- or F- actin may thus be regulated by the local concentrations of the plasma actin-binding proteins gelsolin and DBP. Together, these proteins inhibit platelet aggregation in a manner that can be explained by their effects on actin's filament structure and the accessibility of its bound ADP. Depletion of DBP or gelsolin may allow actin released from injured tissues to stimulate purinergic receptors on platelets, and perhaps other cells, via its bound adenine nucleotides.
Article
A 58-year-old woman with debilitating ankylosing spondylitis who was born to consanguineous parents was found to have an apparent severe vitamin D deficiency that did not respond to supplementation. Liquid chromatography–tandem mass spectrometry showed the absence of circulating vitamin D–binding protein, and chromosomal microarray confirmed a homozygous deletion of the group-specific component (GC) gene that encodes the protein. Congenital absence of vitamin D–binding protein resulted in normocalcemia and a relatively mild disruption of bone metabolism, in this case complicated by severe autoimmune disease. (Funded by the National Institutes of Health and the University of Washington.)
Article
Structured models of ontogenic, phenotypic and functional diversity have been instrumental for a renewed understanding of the biology of immune cells, such as macrophages and lymphoid cells. However, there are no established models that can be used to define the diversity of neutrophils, the most abundant myeloid cells. This lack of an established model is largely due to the uniquely short lives of neutrophils, a consequence of their inability to divide once terminally differentiated, which has been perceived as a roadblock to functional diversity. This perception is rapidly evolving as multiple phenotypic and functional variants of neutrophils have been found, both in homeostatic and disease conditions. In this Opinion article, we present an overview of neutrophil heterogeneity and discuss possible mechanisms of diversification, including genomic regulation. We suggest that neutrophil heterogeneity is an important feature of immune pathophysiology, such that co-option of the mechanisms of diversification by cancer or other disorders contributes to disease progression. © 2019, The Author(s), under exclusive licence to Springer Nature Limited.
Article
Myelopoiesis ensures the steady state of the myeloid cell compartment. Technological advances in fate mapping and genetic engineering, as well as the advent of single cell RNA-sequencing, have highlighted the heterogeneity of the hematopoietic system and revealed new concepts in myeloid cell ontogeny. These technologies are also shedding light on mechanisms of myelopoiesis at homeostasis and at different phases of infection and inflammation, illustrating important feedback loops between affected tissues and the bone marrow. We review these findings here and revisit principles in myelopoiesis in light of the evolving understanding of myeloid cell ontogeny and heterogeneity. We argue for the importance of system-wide evaluation of changes in myelopoiesis and discuss how even after the resolution of inflammation, long-lasting alterations in myelopoiesis may play a role in innate immune memory or trained immunity.
Article
Neutrophils are the first line of defense against bacteria and fungi and help combat parasites and viruses. They are necessary for mammalian life, and their failure to recover after myeloablation is fatal. Neutrophils are short-lived, effective killing machines. Their life span is significantly extended under infectious and inflammatory conditions. Neutrophils take their cues directly from the infectious organism, from tissue macrophages and other elements of the immune system. Here, we review how neutrophils traffic to sites of infection or tissue injury, how they trap and kill bacteria, how they shape innate and adaptive immune responses, and the pathophysiology of monogenic neutrophil disorders.