ArticlePDF AvailableLiterature Review

Vitamin D and immune function in chronic kidney disease

Authors:

Abstract and Figures

The common causes of death in chronic kidney disease (CKD) patients are cardiovascular events and infectious disease. These patients are also predisposed to the development of vitamin D deficiency, which leads to an increased risk of immune dysfunction. Many extra-renal cells possess the capability to produce local active 1,25(OH)2D in an intracrine or paracrine fashion, even without kidney function. Vitamin D affects both the innate and adaptive immune systems. In innate immunity, vitamin D promotes production of cathelicidin and β-defensin 2 and enhances the capacity for autophagy via toll-like receptors activation as well as affects complement concentrations. In adaptive immunity, vitamin D suppresses the maturation of dendritic cells and weakens antigen presentation. Vitamin D also increases T helper (Th) 2 cytokine production and the efficiency of Treg lymphocytes but suppresses the secretion of Th1 and Th17 cytokines. In addition, vitamin D can decrease autoimmune disease activity. Vitamin D has been shown to play an important role in maintaining normal immune function and crosstalk between the innate and adaptive immune systems. Vitamin D deficiency may also contribute to deterioration of immune function and infectious disorders in CKD patients. However, it need more evidence to support the requirements for vitamin D supplementation. Copyright © 2015. Published by Elsevier B.V.
Vitamin-D, innate immunity (anti-infection), and autophagy. The activation of monocyte toll-like receptors (TLR1/TLR2) by pathogen-associated molecular patterns (PAMPs) induces expression of the cytokine interleukin-1 (IL-1) and suppresses expression of the IL-1 receptor antagonist (IL-1RA), thereby enhancing intracrine signaling by IL-1 and increasing nuclear factor кB (NF-кB) activity. The phagocytosis of pathogens increases intracellular concentrations of muramyl dipeptide (MDP), which can then bind to the intracellular pathogenrecognition receptor NOD2. MDP-liganded NOD2 signaling increases NF-кB activity. In addition, the activation of monocyte TLR1/TLR2 by PAMP results in the transcriptional induction of vitamin D receptor (VDR) and 1α-hydroxylase expression. Circulating 25-hydroxyvitamin D [25(OH)D] bound to serum vitamin D-binding protein (DBP) enters monocytes in its free form and is converted to active 1,25-dihydroxyvitamin D [1,25(OH) 2 D] by mitochondrial 1α-hydroxylase, and then binds to VDR. 1,25(OH) 2 D bound to VDR is then able to act as a transcription factor, leading to the induction of cathelicidin and β-defensin 4A expression and the promotion of autophagy by the formation autophagosomes. In concert with 1,25(OH) 2 D bound to VDR, NF-кB also enhances the transcriptional induction of cathelicidin and β-defensin 4A. In the presence of increased cathelicidin, immune cells induce the activity of NOD2/CARD15-β-defensin 2, autophagy-related protein 5 (ATG5), and BECLIN1, then induces autophagy. Antibacterial cathelicidin, β-defensin 4A, and the maturation of autophagosomes then cooperate to enhance bacterial killing. Cytoplasm SNARE proteins mediate fusion between autophagosomes and lysosomes, and various lysosomal enzymes further hydrolyze proteins, lipids, and nucleic acids. The digestive nutrients may be recycled and utilized by the cells. The net efficacy of such a response is highly dependent on vitamin D status, the availability of circulating 25(OH)D for intracrine conversion to active 1,25(OH) 2 D by the enzyme 1α-hydroxylase.
… 
Content may be subject to copyright.
This article appeared in a journal published by Elsevier. The attached
copy is furnished to the author for internal non-commercial research
and education use, including for instruction at the authors institution
and sharing with colleagues.
Other uses, including reproduction and distribution, or selling or
licensing copies, or posting to personal, institutional or third party
websites are prohibited.
In most cases authors are permitted to post their version of the
article (e.g. in Word or Tex form) to their personal website or
institutional repository. Authors requiring further information
regarding Elsevier’s archiving and manuscript policies are
encouraged to visit:
http://www.elsevier.com/authorsrights
Author's personal copy
Invited critical review
Vitamin D and immune function in chronic kidney disease
Wen-Chih Liu
a,b
, Cai-Mei Zheng
a,c
, Chien-Lin Lu
a,d
,Yuh-FengLin
a,c
, Jia-Fwu Shyu
e
,
Chia-Chao Wu
f,
⁎⁎
,1
, Kuo-Cheng Lu
g,
,1
a
Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, No.250, Wuxing Street, Taipei 110, Taiwan
b
Division of Nephrology, Department of Internal Medicine, Yonghe Cardinal Tien Hospital, No.80, Zhongxing St., Yonghe Dist., New Taipei City 234, Taiwan
c
Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, No.291, Zhongzheng Rd., Zhonghe Dist., New Taipei City 235, Taiwan
d
Division of Nephrology, Department of Medicine, Shin-Kong Wu Ho-Su Memorial Hospital, No.95, Wen Chang Road, Shih Lin Dist., Taipei 111, Taiwan
e
Department of Biology and Anatomy, National Defense Medical Center, No.161, Sec. 6, Minquan E. Rd., Neihu Dist., Taipei 114, Taiwan
f
Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, No.325, Sec. 2, Cheng-Kung Rd., Neihu Dist., Taipei 114, Taiwan
g
Department of Medicine, Cardinal Tien Hospital, School of Medicine, Fu Jen Catholic University, No.362, Chung-Cheng Rd, Hsin-Tien Dist., New TaipeiCity 231, Taiwan
abstractarticle info
Article history:
Received 30 June 2015
Received in revised form 13 August 2015
Accepted 14 August 2015
Available online 17 August 2015
Keywords:
Chronic kidney disease
Vitamin D
Innate immunity
Adaptive immunity
The common causes of death in chronic kidney disease (CKD) patients are cardiovascular events and infectious
disease. These patients are also predisposed to the development of vitamin D deciency, which leads to an
increased risk of immune dysfunction. Many extra-renal cells possess the capability to produce local active
1,25(OH)
2
D in an intracrineor paracrine fashion,even without kidney function. Vitamin D affects both the innate
and adaptive immune systems. In innate immunity, vitamin D promotes production of cathelicidin and
β-defensin 2 and enhances the capacity for autophagy via toll-like receptor activation as well as affects
complement concentrations. In adaptive immunity, vitamin D suppresses the maturation of dendritic cells and
weakens antigen presentation. Vitamin D also increases T helper (Th) 2 cytokine production and the efciency
of Treg lymphocytes but suppresses the secretion of Th1 and Th17 cytokines. In addition, vitamin D can decrease
autoimmune disease activity. Vitamin D has been shown to play an important role in maintaining normal
immune function and crosstalk between the innate and adaptive immune systems. Vitamin D deciency may
also contribute to deterioration of immune function and infectious disorders in CKD patients. However, it
needs more evidence to support the requirements for vitamin D supplementation.
© 2015 Elsevier B.V. All rights reserved.
Contents
1. Introduction.............................................................. 136
2. VitaminDmetabolism,function,andregulation.............................................. 136
2.1. VitaminDmetabolism...................................................... 136
2.2. VitaminDfunction ....................................................... 136
2.3. VitaminDregulation ...................................................... 136
2.3.1. VitaminDregulationinnormalsubjects.......................................... 136
2.3.2. VitaminDregulationinCKD............................................... 137
3. VitaminDandimmuneregulation.................................................... 138
4. InnateimmuneresponsesandvitaminDmetabolism ........................................... 138
4.1. VitaminDpromotesinnateimmunity............................................... 138
4.2. VitaminDandantimicrobialpeptides............................................... 138
4.2.1. Cathelicidin ...................................................... 138
4.2.2. Beta-defensins ..................................................... 138
4.3. VitaminDandautophagy .................................................... 139
Clinica Chimica Acta 450 (2015) 135144
Correspondence to: K-C. Lu, Division of Nephrology, Department of Medicine, CardinalTien Hospital, School of Medicine, Fu-Jen Catholic University, No.362, Chung-Cheng Rd, Hsin-
Tien Dist., New Taipei City 231, Taiwan.
⁎⁎ Corresponding author.
E-mail addresses: wucc@ndmctsgh.edu.tw (C.-C. Wu), kuochenglu @gmail.c om (K.-C. Lu).
1
These authors have contributed equally to this work.
http://dx.doi.org/10.1016/j.cca.2015.08.011
0009-8981/© 2015 Elsevier B.V. All rights reserved.
Contents lists available at ScienceDirect
Clinica Chimica Acta
journal homepage: www.elsevier.com/locate/clinchim
Author's personal copy
5. VitaminDandadaptiveimmunity.................................................... 139
5.1. VitaminDandantigenpresentationcells.............................................. 139
5.2. VitaminDandTcellproliferationandactivation .......................................... 140
5.3. VitaminDandTregulatorycells(Tregcells) ............................................ 140
5.4. VitaminDandB-cellfunction................................................... 141
6. ImmunedysfunctionandroleofvitaminDinCKD............................................. 141
6.1. ImmunedysfunctioninCKD ................................................... 141
6.2. RoleofvitaminDinimmunedysfunction(crosstalkbetweeninnateandadaptiveimmunity)...................... 141
7. Conclusions .............................................................. 141
Conictofinterests ............................................................. 141
References ................................................................. 142
1. Introduction
Chronic kidney disease (CKD) patients have chronic inammation,
which is the most important reason for the high morbidity and mortal-
ity associated with CKD [1]. Despite well-established treatments for
CKD, these patients still have a high incidence of cardiovascular and in-
fectious morbidities [1,2]. Mounting evidence indicates the relationship
between cardiovascular complications, infection, and abnormalities in
immunity [36]. Due to impaired renal function and 1α-hydroxylase
deciency, advanced CKD and end-stage renal disease (ESRD) patients
generally lack both 25-hydroxyvitamin D [25(OH)D; calcidiol] and
1,25-dihydroxyvitamin D [1,25(OH)
2
D; calcitriol], which leads to an in-
creased risk of developing infectious complications and cardiovascular
disease. Many factors are involved in the deterioration of immunity in
CKD patients, including anemia, chronic kidney disease mineral
bone disorder (CKDMBD), poor nutrition, chronic inammation, and
dialysis sequelae [1,7,8]. Both the innate and adaptive immune systems
are clearly impaired in CKD patients.
In innate immunity, when pathogen-associated molecular patterns
(PAMPs) stimulate toll-like receptors (TLRs), a kind of pattern recogni-
tion receptor (PRR), on monocytes/macrophages, vitamin D receptor
(VDR) and the expression of 1α-hydroxylase increases. Consequently,
this situation increases the conversion of 25(OH)D to 1,25(OH)
2
D
within monocytes/macrophages in an intracrine manner, producing
cathelicidin and β-defensin to enhance the disinfectant effects of mac-
rophages [9,10] and promote autophagy of intracellular microbes [11].
In contrast, adaptive immunity is set up by antigen presenting cells
(APCs), such as dendritic cells (DCs) and macrophages, which present an-
tigens to T and B lymphocytes. Vitamin D also has an inhibitory effect on
the adaptive immune system by regulating the function of APCs to induce
T lymphocyte activation, proliferation, and cytokine secretion [1214].
However, a reciprocal relationship exists between innate immunity
and adaptive immunity. Interestingly, vitamin D plays a role in the
crosstalk between innate and adaptive immunity because it inuences
DCs. This review focuses on the inuence of vitamin D on innate/
adaptive immunity in CKD patients and discusses the use of vitamin D
to improve immunity in CKD patients.
2. Vitamin D metabolism, function, and regulation
2.1. Vitamin D metabolism
The sources of vitamin D are UVB-dependent endogenous production,
nutritional sources, and supplements. In the skin, 7-dehydrocholesterol
(7-DHC) changes into pre-vitamin D3 when exposed to enough ultravio-
let radiation B (UVB), and vitamin D
3
(cholecalciferol) are easily synthe-
sized with heat [15]. Dietary vitamin D can be divided into plant vitamin
D
2
(ergocalciferol) and animal vitamin D
3
(cholecalciferol). Ergocalciferol
and cholecalciferol are metabolized in the liver by the enzyme vitamin
D-25-hydroxylase and converted to 25-hydroxyvitamin D [25(OH)D]
(calcidiol) as 25(OH)D
2
and 25(OH)D
3
. Clinically, 25(OH)D is the main
circulating form and determines vitamin D status.
When 25(OH)D combines with vitamin D binding protein (DBP), the
resulting complex undergoes glomerular ltration in the kidney. Entry
of the ltered 25(OH)D-DBP compound into proximal renal tubular
cells is facilitated by megalin receptor-mediated endocytosis [16].
Megalin, which is expressed in the renal proximal tubule, is a
multiligand receptor that facilitates the uptake of extracellular ligands.
25(OH)D is delivered to enzyme 1α-hydroxylase by intracellular vita-
min D-binding protein 3 (IDBP3) and changes to active form.The mito-
chondrial 24-hydroxylase protein initiates the degradation of 25(OH)D
and 1,25(OH)
2
D by hydroxylation of the side chain to form the inactive
blood metabolite 24,25(OH)
2
D and calcitroic acid [17,18]. The half-life
of Vitamin D is different depending on its form; the half-life of nutrition-
al vitamin D is 60 days, 25(OH)D is 23 weeks, and 1,25(OH)
2
Dis
812h[19]. In the circulation, 1,25(OH)
2
D has 1000 times stronger
afnity for the specic vitamin D receptor than 25(OH)D in serum, it
delivers a higher biologic activity and a shorter half-life [20]. However,
the level of 25(OH)D is 1000 times greater than the level of
1,25(OH)
2
D[21].
Interestingly, vitamin D is also metabolized to active 1,25(OH)
2
D
without kidney function, as numerous extra-renal cells possess their
own 1α-hydroxylation capability [10,2227]. Circulating 25(OH)D is
converted to 1,25(OH)
2
D in the extra-renal tissues and acts locally in
both an autocrine and paracrine manner. The percentage of extra-
renal 1α-hydroxylation may be higher in CKD [27,28]. Furthermore,
VDR is found not only in many traditional organs, such as bone and
the kidneys, but also in the immune system [2931]. Rationally, the
extra-skeletal functions of vitamin D consist of protecting renal func-
tion, protecting the cardiovascular system, regulating the immune sys-
tem, and preventing cancer, among others.
2.2. Vitamin D function
The traditional effects of 1,25(OH)
2
D are related to b one metabolism
via the regulation of blood calcium, phosphate, and parathyroid hor-
mone concentrations [32]. In addition to the management of calcium
and phosphorus homeostasis, 1,25(OH)
2
D has many benecial effects
due to non-calciotropic actions. Several studies have shown that
1,25(OH)
2
D induces anti-cell differentiation, inhibits cell proliferative
activity, and regulates adaptive immunity by decreasing the production
of inammatory cytokines [12,22,3335].
In addition, stimulation of TLRs on monocytes/macrophages by
pathogens, such as Mycobacterium tuberculosis and other bacteria,
leads to increased expression of VDR in macrophages/monocytes [36].
Therefore, 1,25(OH)
2
D enhances the production of cathelicidin to pro-
mote the disinfectant effects of macrophages and monocytes [9,10].
2.3. Vitamin D regulation
2.3.1. Vitamin D regulation in normal subjects
Vitamin D regulates organ functions by binding to its receptor on the
cell surface. In the kidneys, vitamin D protects the kidney function
by decreasing the progression of renal brosis, inammation, and
136 W.-C. Liu et al. / Clinica Chimica Acta 450 (2015) 135144
Author's personal copy
proteinuria [37]. In bone mineralization, 1,25(OH)
2
D is recognized by its
receptor on osteoblasts to promote osteoblast maturation and increases
calcium and phosphate absorption in the small intestine by combining
with the vitamin D receptorretinoic acid x-receptor complex (VDR
RXR) to augment the expression of the epithelial calcium channel [36].
In addition, 1,25(OH)
2
D diminishes the synthesis and secretion of
parathyroid hormone (PTH) in the parathyroid gland.
2.3.2. Vitamin D regulation in CKD
Advanced CKD and ESRD patients are prone to deciencies in both
25(OH)D and 1,25(OH)
2
D. When renal function worsens, serum
1,25(OH)
2
D levels decline, even before any changes in serum calcium
or phosphorus concentrations. The ideal levels of 25(OH)D in the
serum is unknown but may be slightly higher than 30 ng/ml in CKD
patients [30] for extra-renal production of 1,25(OH)
2
D and regulation
of PTH secretion. Vitamin D deciency can occur in CKD patients for a
number of reasons.
2.3.2.1. Decreased 25(OH)D (calcidiol) in CKD. Because renal mass and
the glomerular ltration rate (GFR) are decreased in CKD patients, lim-
ited 25(OH)D enters the renal tubules and less 25(OH)D is taken up. In
addition, damaged kidneys have decreased megalin expression, which
impairs 25(OH)D reabsorption. Furthermore, albuminuria will occupy
the megalin route and fewer receptors will be available to reabsorb
25(OH)D-DBP. In addition, as a consequence of proteinuria, proximal
tubular cells are damaged and express less megalin [38] (Fig. 1).
Therefore, CKD reduces 25(OH)D resorption and results in the
formation of less 1,25(OH)
2
D in a substrate-dependent process [39].
2.3.2.2. Decreased 1α-hydroxylase activity and increased 24-hydroxylase
activity. Renal tubular cells contain two enzymes related to vitamin D
metabolism: 1α-hydroxylase and 24-hydroxylase. These enzymes can
hydroxylate 25(OH)D to either 1,25(OH)
2
Dor24,25(OH)
2
D, an inactive
metabolite. CKD, diabetic, increased FGF-23, and the use of active vita-
min D decrease the activity of 1α-hydroxylase and 25-hydroxylase
Fig. 1. Vitamin D metabolism and its regulations. Very few foods contain vitamin D and synthesis of vitamin D (specically cholecalciferol) in the skin is the major natural source of the
vitamin. Dermal synthesis of vitamin D from cholesterol is dependent on sun exposure. Vitamin D obtained from the diet or dermal synthesis is biologically inactive. The activation of
vitamin D requires enzymat ic conversion (hydroxylation) in the liver (25-hydr oxylase) and kidney (1α-hy droxylase). The mitochond rial protein (24-hydroxylase) initiates the
degradation of 25-hydroxyvitamin D [25(OH)D] and 1,25-dihydroxyvitamin D [1,25(OH)
2
D] by hydroxylation of the side chain to form calcitroic acids. In CKD, diabetic, increased
broblastgrowth factor 23 (FGF-23), and the use of active vitamin D decreases the activity of 25-hydroxylase and 1α-hydroxylase and increasesthe activity of 24-hydroxylase, resulting
in decreased endogenous25(OH)D and 1,25(OH)
2
D production and increased25(OH)D and 1,25(OH)
2
D degradation. In CKD patientswith secondary hyperparathyroidism, a large dose of
activevitamin D may aggravate25(OH)D deciency, which may also deprivethe need for 25(OH)D in othertissues or organs (skin,prostate, colon, brain,breast, lung, placenta, osteoblast,
parathyroid gland, pancreas, muscle, monocyte, T/B cells, etc.).
137W.-C. Liu et al. / Clinica Chimica Acta 450 (2015) 135144
Author's personal copy
and increase the activity of 24-hydroxylase, resulting in a decrease in
endogenous 25(OH)D and 1,25(OH)
2
D production and increased
25(OH)D and 1,25(OH)
2
Ddegradation[40,41].
2.3.2.3. Inuence of phosphaturic hormones. In CKD, FGF-23, a
phosphaturic hormone, is secreted to maintain normal serum phos-
phate homeostasis. As kidney function deteriorates, FGF-23 markedly
increases in individuals with CKD [42]. FGF-23 inhibits renal production
of 1,25(OH)
2
Dbydecreasing1α-hydroxylase activity in renal proximal
tubules and simultaneously increasing the expression of 24-hydroxylase
and production of 24,25(OH)
2
D[43]. Therefore, FGF-23 decreases the
25(OH)D concentration.
2.3.2.4. Overusage of 1,25(OH)
2
D. 1,25(OH)
2
D is the most common drug
prescribed in CKD patients with secondary hyperparathyroidism.
Because 1,25(OH)
2
D, the end product of vitamin D, feedback inhibits
1α-hydroxylase and 25-hydroxylase, a large dose of 1,25(OH)
2
D may
aggravate 25(OH)D deciency, which may decrease the providing of
25(OH)D in other tissues or organs (e.g., colon, brain, breast, osteo-
blasts, parathyroid gland, monocytes, immune cells) [44]. In addition,
the activity of 1α-hydroxylase and concentration of 25(OH)D have a
substrate-dependent relationship in CKD patients; the higher concen-
tration of 25(OH)D, the stronger the 1α-hydroxylase activity, and vice
versa [45].
3. Vitamin D and immune regulation
More than one hundred years ago, vitamin D was initially used to
treat infections and cod liver oil successfully helped tuberculosis [1].Im-
mune cells carry VDR and 1α-hydroxylase, which profoundly inuences
human health [36,46]. The immune system produces the active metab-
olite 1,25(OH)
2
D through local synthesis and heightens immunomodu-
latory properties [47,48]. Increasing evidence indicates that vitamin D
deciency, as in CKD, not only causes dysregulation of the innate and
adaptive immune systems, but also promotes microinammation [49].
In addition, epidemiological experiments have found that vitamin Dde-
ciency is closely related to autoimmune and infectious diseases
[5053]. When serum vitamin D levels are adequate, the activation of
innate immunity is immediately enhanced when microbial pathogens
are presented, and the benets of the adaptive immune response
prevent various autoimmune diseases and transplant rejection [1].
In addition, a cross-sectional analysis reportedthat severely ill septic
patients have signicantly low serum 25(OH)D levels. The levels were
related to diminished concentrations of antimicrobial peptides, such
as cathelicidin and β-defensin 2[5456]. Another randomized con-
trolled trial demonstrated that daily intake of 4000 IU cholecalciferol
over one year meaningfully reduces complications of infection,
pathogens in the nasal uid, and antibiotic use [57]. Therefore, vitamin
D would upregulate antimicrobial peptide levels and be essential in
infection control.
4. Innate immune responses and vitamin D metabolism
4.1. Vitamin D promotes innate immunity
Innate immunity identies PAMPs through TLRs and initiates
defense mechanisms in response to microorganisms [58]. Recently,
1,25(OH)
2
D was shown to enhance the effects of macrophages and
monocytes against pathogens by stimulating the differentiation of
monocytes into mature phagocytic macrophages [5961]. During
infection, macrophages/monocytes are exposed to PAMPs, a small
molecular motif preserved within a class of microbes that activates
TLR 1/2 heterodimer. 1α-Hydroxylase activity and VDR expression are
then upregulated to produce 1,25(OH)
2
D[10,55]. Lipopolysaccharide
also induces TLR4, interferon-γ(IFN-γ), and CD14 activity to increase
1α-hydroxylase expression [1,62] (Fig. 2).
When serum 25(OH)D levels are N30 ng/ml (75 nmol/l), it can con-
vert to its active form, 1,25(OH)
2
D, via 1α-hydroxylase in macrophages
in an intracrine or autocrine manner [36].1,25(OH)
2
D enters the nucle-
us by binding VDR and complexes with retinoid X receptor (RXR),
directly signaling the transcription of cathelicidin and β-defensin 2
[33,5456,63]. Both of these peptides cleave microbial membranes
and promote innate immunity in response to infectious agents, such
as bacteria, viruses, and fungi, in humans [6466].
Compared to people with adequate serum levels of vitamin D, mac-
rophage function in vitamin D-decient patients may deteriorate and
lead to a decreased bacterial killing effect [67].However,somestudies
have shown that, in contrast to renal 1α-hydroxylase, local production
of 1α-hydroxylase in macrophages is not inhibited by endocrine
1,25(OH)
2
D and is promoted by immune stimuli, such as interleukin-1
(IL-1) and IFN-γ[68,69].
Since 25(OH)D levels of individuals decide the capability of vitamin
Dtoinuence normal human immunity, vitamin D insufciency may
lead to a poor reaction to infection or innate immunity. In Swiss,
Sakem et al. found that almost two-thirds of healthy and older people
showed of Vitamin D insufciency. They also noticed that low levels of
25(OH)D were directly related to levels of IgG2 and complement
component C4, in contrast, which were opposite associated with levels
of IgG1 and IgA and complement component C3. In other words, the
higher level of 25(OH)D will accompany with increasing C4 concentra-
tion but decreasing C3 concentration [70].
4.2. Vitamin D and antimicrobial peptides
4.2.1. Cathelicidin
Cathelicidin, the antimicrobial protein found in lysosomes, macro-
phages, and polymorphonuclear leukocytes, damages microbial mem-
branes and is increased in defense against infection [64].InCKD
patients, low 1,25(OH)
2
D levels have been related to elevated mortality
rates caused by severe infections [71]. Thus, the vitamin D concentration
increases antimicrobial peptide levels and may be essential in the treat-
ment of infections. A study of African-Americans with lower levels of
25(OH)D found that, after providing 25(OH)D supplements, adequate
serum concentrations were achieved, rescuing TLR activation and in-
duction of cathelicidin mRNA [10]. Previous studies showed that
cathelicidin can be upregulated by 1,25(OH)
2
D via vitamin D response
element (VDRE) complexes [54]. Other experiments demonstrated
that VDR can increase cathelicidin transcription and 1,25(OH)
2
D can
build up cathelicidin levels in several types of cells, including
monocytes, macrophages, and neutrophils [54,72]. Liu et al. published
two important studies: the rst one demonstrated that decreased
cathelicidin levels can diminish the monocyte capacity for killing
M. tuberculosis, and the second demonstrated that vitamin D supple-
mentation can enhance cathelicidin levels and promote antibacterial
activity [10].
4.2.2. Beta-defensins
Vitamin D binding to VDR can upregulate the expression of the
β-defensin 4A (DEFB4A) gene, which encodes the antibacterial
β-defensin 2 protein, during the innate immune response [54,55].
Compared to cathelicidin, vitamin D has less in uence on DEFB4 expres-
sion because the functional ability of DEFB4VDRE requires other stim-
uli to increase transcriptional activity [55]. Gastrointestinal epithelial
interleukin-1 (IL-1b) also enhances DEFB4 expression [73].Inaddition,
DEFB4 is induced by 1,25(OH)
2
D through nucleotide-binding oligomer-
ization domain 2 (NOD2), an intracellular PRR that binds muramyl
dipeptide (MDP), a cell membrane product of bacteria [74]. The activa-
tion of immune cells induces the expression of IL-1 and suppresses ex-
pression of the IL-1 receptor antagonist, thereby enhancing intracrine
signaling by IL-1 and increasing nuclear factor кB(NF-кB) activity [75].
The NF-кB pathway also upregulates NOD2 signals to enhance DEFB4
138 W.-C. Liu et al. / Clinica Chimica Acta 450 (2015) 135144
Author's personal copy
activity in a similar manner [76]. Therefore, vitamin D produces DEFB4
throughNOD2activationandNF-кB stimulation [77].
4.3. Vitamin D and autophagy
In addition to bacterial killing agents, there are other essential fac-
tors that inuence innate immune responses to pathogen invasion.
The cellular environment is also an important bactericidal location. It
is advantageous to kill pathogens when the merger of phagosomes
with lysosomes creates a phagolysosomal environment. Although the
mechanism is still unclear, autophagy, another aspect of cellular
immune function, may enhance the procedure of phagocytosed
pathogens.
In the presence of increased cathelicidin levels, immune cells induce
the activity of NOD2/CARD15-β-defensin 2, autophagy-related protein
5 (ATG5), and BECLIN1, which induce autophagy [76,78]. Autophagy is
not only the elimination of materials, but also acts as a dynamic
recycling system that yields new components and energy for cellular
renovation and homeostasis. Autophagy increases in response to sever-
al stimuli, such as infection and starvation [79]. Therefore, autophagy is
an important defense mechanism of macrophages against intracellular
pathogens [11]. Antibacterial cathelicidin, β-defensin 4A, and matura-
tion of autophagosomes cooperate to enhance bacterial killing. In addi-
tion, cytoplasm SNARE proteins mediate the fusion of autophagosomes
and lysosomes, and various lysosomal enzymes further hydrolyze
proteins, lipids, and nucleic acids. The digestive nutrients may be
recycled and utilized by the cells. The net efcacy of such a response
is highly dependent on vitamin D status, the availability of circulating
25D for intracrine conversion to active 1,25D by the enzyme
1α-hydroxylase [65].
5. Vitamin D and adaptive immunity
Decades ago, human lymphocytes were observed to present VDR
associating with vitamin D in extramineral reactions [80].Evidence
indicates that VDR are presented in both activated T cells and B cells
[81,82] but not inactive cells. Thus, vitamin D plays a functional role in
modulating adaptive immunity. This process is set up by APCs, such as
DCs and macrophages, which present antigens to T lymphocytes and B
lymphocytes, leading to the production of antibodies and the regulation
of immunological memory [12].
5.1. Vitamin D and antigen presentation cells
The important distinction between innate and adaptive immunity is
the existence of APCs [77]. DCs, professional APCs, can express MHC
class II and have the necessary costimulatory signals to activate CD4
+
T cells (helper T-cells). In contrast, most somatic cells present MHC
class I molecules and act as the targets of CD8
+
T cells (cytotoxic
T-cells). In immature DCs, TLRs are stimulated by microbiological or
bacterial lipopolysaccharides to enhance DC maturity. Mature DCs
travel to lymph nodes to interact with T and B cells under the inuence
of cytokines and chemokines.
In VDR knockout mice, Grifn et al. showed the important role of
VDR in DC maturation responses to vitamin D metabolites [83]. Later,
Piemonti et al. found that vitamin D leads to DC immaturity and results
Fig. 2. Vitamin-D, innate immunity (anti-infection), and autophagy. The activation of monocyte toll-like receptors (TLR1/TLR2) by pathogen-associated molecular patterns (PAMPs)
induces expression of the cytokineinterleukin-1 (IL-1) and suppresses expression of the IL-1 receptor antagonist (IL-1RA), thereby enhancing intracrine signaling by IL-1 and increasing
nuclear factor кB(NF-кB) activity. The phagocytosisof pathogens increases intracellular concentrations of muramyl dipeptide (MDP), which can then bind to the intracellularpathogen-
recognition receptor NOD2. MDP-liganded NOD2 signaling increases NF-кBactivity. In addition,the activation of monocyte TLR1/TLR2 by PAMP results in the transcriptional induction of
vitaminD receptor (VDR)and 1α-hydroxylase expression.Circulating 25-hydroxyvitaminD [25(OH)D] bound to serumvitamin D-bindingprotein (DBP) entersmonocytes in its freeform
and is convertedto active 1,25-dihydroxyvitaminD [1,25(OH)
2
D] by mitochondrial 1α-hydroxylase, and then bindsto VDR. 1,25(OH)
2
D bound to VDR is then ableto act as a transcription
factor,leading to the induction of cathelicidinand β-defensin4A expression andthe promotion of autophagy by the formation autophagosomes. In concertwith 1,25(OH)
2
D bound to VDR,
NF-кB also enhances the transcriptional induction of cathelicidin and β-defensin 4A. In the presence of increased cathelicidin, immune cells induce the activity of NOD2/CARD15-β-
defensin 2, autophagy-related protein 5 (ATG5), and BECLIN1, then induces autophagy. Antibacterial cathelicidin, β-defensin 4A, and the maturation of autophagosomes then cooperate
to enhance bacterial killing. Cytoplasm SNARE proteins mediate fusion between autophagosomes and lysosomes, and various lysosomal enzymes further hydrolyze proteins, lipids, and
nucleic acids. The digestive nutrients may be recycled and utilized by the cells. The net efcacy of such a response is highly dependent on vitamin D status, the availability of circulating
25(OH)D for intracrine conversion to active 1,25(OH)
2
D by the enzyme 1α-hydroxylase.
139W.-C. Liu et al. / Clinica Chimica Acta 450 (2015) 135144
Author's personal copy
in failed antigen presentation [8486].NF-кB signaling plays an impor-
tant role in the inhibitory effect of vitamin D on DC maturation [87,88].
There are two major reasons for immature DCs in vitamin D metab-
olism. First, 25(OH)D or 1,25(OH)
2
D suppresses the maturation of DCs
because of decreased expression of the co-stimulatory markers HLA-
DR, CD40, CD80, and CD86, which are necessary for antigen presenta-
tion and T cell proliferation [8991]. 1,25(OH)
2
D also decreases the
level of IL 12, which can enhance the stimulation of T helper 1 (TH1)
cell development [92]. Second, the other pivotal characteristics of
vitamin D metabolism in DCs is that there is a relevant weakening of
the expression of VDR and ability of 1,25(OH)
2
D to bind the receptor
when the expression of 1α-hydroxylase and synthesis of 1,25(OH)
2
D
increases [91,93]. Because mature DCs have plenty of 1α-hydroxylase
to convert 25(OH)D to 1,25(OH)
2
D, which inuences immature DCs
that express higher levels of the VDR than their mature counterparts.
In this paracrine mechanism, 1,25(OH)
2
D prevents the maturation of
DCs, thereby reducing antigen presentation and improving the T cell
tolerogenic immune response [77], preventing over-amplication of
adaptive immune responses [94].
5.2. Vitamin D and T cell proliferation and activation
1,25(OH)
2
D plays a key role in the proliferation and activation of T
cells. Vitamin D leans toward a tolerogenic immune status instead of
inammation and promotes a T cell shift from Th1 to Th2 [95].This
situation potentially damages tissue related to Th1 cellular immune
responses. Usually, vitamin D increases anti-inammatory Th2 cytokine
(i.e., IL-3, IL-4, IL-5, and IL-10) production [96] and the efciency of Treg
lymphocytes but suppresses the secretion of Th1 cytokines (i.e., IL-2,
IFN-γ, and TNF-α), which are potent stimulators of inammation [90,
97,98] (Fig. 3). There are several mechanisms by which serum vitamin
Dinuences T cell function: endocrine, paracrine, and intracrine. Firstly,
systemic endocrine 1,25(OH)
2
D directly affects T cells. Secondly,
25(OH)D is converted to local 1,25(OH)
2
D within DCs acting on T cells
in a paracrine fashion. Thirdly, either the synthesis of 1,25(OH)
2
Din
an intracrine manner in DCs indirectly affects antigen presentation to
T cells or 25(OH)D directly converts to 1,25(OH)
2
DinTcellsinan
intracrine manner [99].
Vitamin D also inhibits the development of Th17 (i.e., IL-17, IL-21),
which plays an essential role in the occurrence of autoimmune diseases
associated with tissue damage, inammation, and host-graft rejection
[100]. Penna et al. demonstrated in an in vitro study that treatment of
non-obese diabetes (NOD) mice, a model of susceptible autoimmune
disease, with a synthetic analog of 1,25(OH)
2
D could reduce IL-17 ex-
pression [101]. Thus, vitamin D directly inuences T cell proliferation
and cytokine production [102]. Patients on chronic hemodialysis (HD)
have impaired cellular and humoral immunity, and we have demon-
strated that Th2 differentiation correlates with age and serum
25(OH)D levels in patients [103]. We found that higher phosphate and
iPTH levels and longer dialysis duration increase Th17 cell differentia-
tion, especially in non-diabetic chronic HD patients [104].
5.3. Vitamin D and T regulatory cells (Treg cells)
Treg cells exhibit anti-inammatory effects and reduce autoimmune
disorders by releasing cytokines IL-10 and TGF-β[105]. The suppression
of DC maturation by 1,25(OH)
2
D weakens the adaptive T lymphocyte
responses and augments immunosuppression affecting by Treg cells
Fig. 3. Mechanisms for adaptive immune responses to vitamin D. Dendritic cells (DCs) expressing 1α-hydroxylase and the vitamin D receptor (VDR) can utilize circulating
25-hydroxyvitaminD [25(OH)D] for intracrine responses vialocalized conversionto active 1,25-dihydroxyvitaminD [1,25(OH)
2
D]. In DCs, intracrine synthesisof 1,25(OH)
2
DinhibitsDC
maturation, thereby modulating helper T (Th) cellfunction. Th responses to 25(OH)D may alsobe mediatedin a paracrinefashion,with DC-generated 1,25(OH)
2
D actingon VDR-expressing
Th cells. VDR-expressing Th cells are also potential targets for systemic 1,25(OH)
2
D (endocrine effect). The action of vitaminD on DCs stimulates effector CD4
+
cells todifferentiate into one
of the four types of Th cells. Activated T cells also expressVDR. Under normal conditions vitaminD increases Th2 cytokines(i.e., IL-10) and the efciency of regulatory T (Treg) lymphocytes.
Vitamin D inhibits the development of Th1 cells, which are associated with the cellular immune response. In addition, vitamin D promotes Th2 cells associated with humoral (antibody)
mediated immunity. Thus, vitamin D promotes the T cell shift from Th1 to Th2 function. Vitamin D also inhibits the development of Th17 cells, which play an essential role in combating
certain pathogens and are linked to tissue damage and inammation. The fourth group of CD4
+
T cells, Treg cells, exerts suppressor function responses to vitamin D.
140 W.-C. Liu et al. / Clinica Chimica Acta 450 (2015) 135144
Author's personal copy
[106]. By inhibiting the maturation of DCs and increasing expression of
DC cytokines, such as CCL22, 1,25(OH)
2
D has the potential to suppress
Th1 cells and induce CD4
+
/CD25
+
Treg cells [107], which are essential
for the induction of tolerogenic immune responses [108].Theinuence
of vitaminD on Tregs can be directthrough endocrinesystemic calcitriol
effects or intracrine conversion of 25(OH)D to 1,25(OH)
2
DbyTregs
themselves, or indirect via APCs remaining in the immature status
while providing vitamin D supplementation, resulting in less antigen
expression [109]. Prietl et al. demonstrated that high doses of cholecal-
ciferol in the healthy population can signicantly increase the percent-
age of Tregs in peripheral blood [110]. Ardalan et al. also found that
supplementing calcitriol upregulates the population of CD4
+
CD25
+
T
cells in renal transplant recipients [111].
5.4. Vitamin D and B-cell function
1,25(OH)
2
D results in reduced proliferation and differentiation of B
lymphocytes and the production of immunoglobulin, leading to apopto-
sis because VDR can be expressed at higher levels in active B lympho-
cytes compared to resting B lymphocytes [112]. These effects are
probably indirectly mediated by T cells and macrophages [95,113].
This retardation of β-cell precursors into plasma cells is important in au-
toimmune diseases because plasma cells produce autoantibodies that
play a key role in the mechanisms underlying autoimmune disease
such as systemic lupus erythematosus (SLE) [32] and Crohn's disease
[114].
6. Immune dysfunction and role of vitamin D in CKD
6.1. Immune dysfunction in CKD
CKD patients usually have obvious immune dysregulation compared
to the general population. Due to the immune dysregulation, CKD pa-
tients have vascular calcication, atherosclerosis, impaired glucose tol-
erance, and hypercytokinemia [115,116]. Thus, they not only have a
higher rate of infection and malignancy, but also increased incidence
of cardiovascular events [117119]. Many studies have shown that
both the innate and adaptive immune systems are impaired in patients
with advanced CKD and ESRD [4,5]. In CKD patients,immunity is usually
pre-activated to overproduce pro-inammatory cytokines, such as TNF-
α, IL-1, and IL-6 [120,121], and the clearance of these hypercytokines is
impaired [122,123].
It is about 90% of health people who received hepatitis B vaccines
will achieve sufcient titers of anti-HBs antibodies but just 5060% of
ESRD patients can obtain enough anti-HBs antibody titers [124]. Many
disturbances of immunity in CKD patients result in this impaired anti-
body response [117]. Zitt et al. conducted a research about anti-HBs an-
tibody titers and serum 25(OH)D levels in CKD 35D patients who
received hepatitis B vaccination, which performed that a lower antibody
titer is related to vitaminD deciency. They assume that rising the levels
of 25(OH)D before hepatitis B vaccination has the possibility to increase
the titers of anti-HBs antibody and strengthen the immune responses
[125].
Furthermore, CKD attenuates the function of both DCs and macro-
phages because co-stimulatory molecules such as CD40, CD80, and
CD86 are impaired [30,126128]. Consequently, CKD leads to a dimin-
ished response to infection and misapplied inammatory response as
in a state of immune dysregulation and sustained inammation [1].In
addition, fewer peripheral CD4
+
Tlymphocytes,CD8
+
Tlymphocytes,
and B lymphocytes are present, but a higher level of soluble B lympho-
cyte markers is found in the blood of CKD patients [129,130]. Therefore,
the immune deterioration in CKD patients is a complex issue. Some
studies have demonstrated that uremic toxins lead to the accumulation
of pro-inammatory cytokines and immunosuppression [3,131].
6.2. Role of vitamin D in immune dysfunction (crosstalk between innate
and adaptive immunity)
DCs are the key connector betweenthe innate and adaptive immune
systems. In the innate response, DCs are activated by PRRs and sequen-
tially initiate the adaptive immune network [132]. Vitamin D plays a
role in the crosstalk between innate and adaptive immunity because it
has an essential inuence on DCs (Fig. 4). In macrophages, 25(OH)D is
converted to local 1,25(OH)
2
D through 1α-hydroxylase, and this pro-
duction induces paracrine responses in monocytes and T or B lympho-
cytes. Activated monocytes further promote the differentiation of
macrophages via the paracrine effects of vitamin D, indicating that vita-
min D can enhance innate immunity. In contrast, vitamin D inhibits the
expression of MHC-II molecules and co-stimulatory molecules [133,
134]. Thus, local 1,25(OH)
2
D is assumed to have immunosuppressive
properties. In addition, the synthesis of vitamin D leads to the attenua-
tion of antigen presentation and enhances a tolerogenic immune
response. Vitamin D suppresses the maturation of DCs and promotes
the apoptosis of mature DCs [85], which inhibits adaptive immunity,
increasing the number of Th2 cells and Treg cells, and decreasing the
number of Th1 and Th17 cells [135].
Additionally, IFN-γ, produced by natural killer (NK) cells and T cells
[136], is a cytokine that is important for innate and adaptive immunity.
In innate immunity, IFN-γincreases the expression of 1α-hydroxylase
to augment local 1,25(OH)
2
D production and enhance macrophage
phagocytosis. T cells express IFN-γthrough the stimulation of IL-15
secreted by DCs [137]. Therefore, a different mechanism in the adaptive
immune system can enrich the innate immune response to pathogens,
as increasing the activity of macrophage 1α-hydroxylase is indepen-
dent of PAMPs [77].
7. Conclusions
In CKD and ESRD patients, there is a tendency for immune dysfunc-
tion and infectious disorders due to vitamin D deciency. Mounting
evidence indicates that the use of vitamin D goes beyond its traditional
roles, such as maintaining mineral homeostasis and dealing with
secondary hyperparathyroidism in CKD. Consequently, vitamin D has
received attention because of its pleiotropic actions in many chronic
diseases. However, it still needs more evidence to support the need for
vitamin D supplementation in improving immune function. Many
studies have established that extra-renal tissues and cells, such as im-
mune cells, contain vitamin D metabolizing enzymes (1-αhydroxylase)
to produce local biologically active forms of vitamin D in an intracrine/
paracrine manner.
Vitamin D promotes innate immune effects and inhibits adaptive
immune effects. In innate immunity, 1,25(OH)
2
D can be synthesized
by inammatory cells during infection. Macrophages recognize PAMPs
via TLRs and increase the production of β-defensin 2 and cathelicidins
to ght against pathogens. In addition, cathelicidins induce the activa-
tion of autophagy, whichis an important defense mechanism of macro-
phages to kill bacteria. In adaptive immunity, vitamin D can promote
the tolerogenic function of Treg cells and decrease autoimmune disease.
Recently, vitamin D has been shown to play an important role in the
crosstalk between the innate and adaptive immune systems. In other
words, vitamin D essentially inuences DCs, which are the interface be-
tween innate and adaptive immunity. Collectively, vitamin D has a very
important role in maintaining normal immune function, and vitamin D
deciency or insufciency inCKD patients could result in a deterioration
of immune function and infectious disorders.
Conict of interests
The authors declare that there is no conict of interests regarding
the publication of this paper.
141W.-C. Liu et al. / Clinica Chimica Acta 450 (2015) 135144
Author's personal copy
References
[1] K.A. Sterling, P. Eftekhari, M. Girndt, P.L. Kimmel, D.S. Raj, The immunoregulatory
function of vitamin D: implications in chronic kidney disease, Nat. Rev. Nephrol.
8 (7) (2012) 403412.
[2] M. Tonelli, N. Wiebe, B. Culleton, A. House, C. Rabbat, M. Fok, et al., Chronic kidney
disease and mortality risk: a systematic review, J. Am. Soc. Nephrol.: JASN 17 (7)
(2006) 20342047.
[3] P.L. Kimmel, T.M. Phillips, S.J. Simmens, R.A. Peterson, K.L. Weihs, S. Alleyne, et al.,
Immunologic function and survival in hemodialysis patients, Kidney Int. 54 (1)
(1998) 236244.
[4] S. Kato, M. Chmielewski, H. Honda, R. Pecoits-Filho, S. Matsuo, Y. Yuzawa, et al.,
Aspects of immune dysfunction in end- stage renal disease, Clin. J. Am. Soc.
Nephrol.: CJASN 3 (5) (2008) 15261533.
[5] B.L. Jaber, Bacterial infections in hemodialysis patients: pathogenesis and prevention,
Kidney Int. 67 (6) (2005) 25082519.
[6] A.E. Stinghen, S . Bucharles, M.C. Riella, R. Pecoits-Filho, Immu ne mechanisms
involved in cardiovascular complications of chronic kidney disease, Blood Purif.
29 (2) (2010) 114120.
[7] C. Tzanno-Martins, E. Futata, V. Jorgetti, A.J. Duarte, Restoration of impaired T-cell
proliferation after parathyroidectomy in hemodialysis patients, Nephron 84 (3)
(2000) 224227.
[8] R. Vanholder, R. De Smet, C. Hsu, P. Vogeleere, S. Ringoir, Uremic toxicity: the
middle molecule hypothesis revisited, Semin. Nephrol. 14 (3) (1994) 205218.
[9] M. Fabri, S. Stenger, D.M. Shin, J.M. Yuk, P.T. Liu, S. Realegeno, et al., Vitamin D is
required for IFN-gamma-mediated antimicrobial activity of human macrophages,
Sci. Transl. Med. 3 (104) (2011) (104ra02).
[10] P.T. Liu, S. Stenger, H. Li, L. Wenzel, B.H. Tan, S.R. Krutzik, et al., Toll-like receptor
triggering of a vitamin D-mediated human antimicrobial response, Science 311
(5768) (2006) 17701773.
[11] V. Deretic, Autop hagy in immunity and cell-autonomous defense against
intracellular microbes, Immunol. Rev. 240 (1) (2011) 92104.
[12] D.D. Bikle, Vitamin D, and immune function: understanding common pathways,
Curr. Osteoporos. Rep. 7 (2) (2009) 5863.
[13] W.F.Rigby, R.J. Noelle, K. Krause, M.W.Fanger, The effectsof 1,25-dihydroxyvitamin
D3 on human T lymphocyte activation and proliferation: a cell cycle analysis , J.
Immunol.135 (4) (1985) 22792286.
[14] A.K. Bhalla, E.P. A mento, B. Serog, L.H. Glimcher, 1,25-Dihydroxyvitamin D3
inhibits antigen-induced T cell activation, J. Immunol. 133 (4) (1984) 17481754.
[15] M.F. Holick, Vitamin D: a D-Lightful health perspective, Nutr. Rev. 66(10 Suppl. 2)
(2008) S182S194.
[16] A. Nykjaer, D. Dragun, D. Walther, H. Vorum, C. Jacobsen, J. Herz, et a l., An
endocytic pathwa y essential for renal uptake and activation of the steroid
25-(OH) vitamin D3, Cell 96 (4) (1999) 507515.
[17] K.S. Chen, H.F. DeLuca, Cloning of the human 1 alpha,25-dihydroxyvitamin D-3
24-hydroxylase gene promoter and identication of two vitamin D-responsive
elements, Biochim. Biophys. Acta 1263 (1) (1995) 19.
[18] Y. Ohyama, K. Ozono, M. Uchida, T. Shinki, S.Kato, T. Suda, et al., Identication of a
vitamin D-responsive element in the 5-anking region of the rat 25-hydroxyvitamin
D3 24-hydroxylase gene, J. Biol. Chem. 269 (14) (1994) 1054510550.
[19] A. Zitterma nn, S.S. Schleith off, S. Frisch, C. Gotting, J. Kuhn, H. Koertke, et al.,
Circulating cal citriol concentr ations and total mo rtality, Clin. Chem. 55 (6)
(2009) 11631170.
[20] S. Mazzaferro, D. Gold smith, T.E. Larsson, Z.A. M assy, M. Cozzolino, Vitam in D
metabolites and/or analogs: which D for which patient? Curr. Vasc. Pharmacol.
12 (2) (2014) 339349.
[21] A. Massart, F.D. Debelle, J. Racape, C. Gervy, C. Husson, M. Dhaene, et al., Biochem-
ical parameters after cholecalciferol repletion in hemodialysis: results from the
VitaDial randomized trial, Am. J. Kidney Dis.: the ofcial journal of the National
Kidney Foundation 64 (5) (2014) 696705.
[22] A.S. Dusso, A.J. Brown, E. Slatopolsky, Vitamin D, Am. J. Physiol. Renal Physiol. 289
(1)(2005)F8F28.
[23] D.L. Andress, Vitamin D, in chronic kidney disease: a systemic role for selective
vitamin D receptor activation, Kidney Int. 69 (1) (2006) 3343.
[24] A.J. Brown, Therapeutic uses of vitamin D analogues, Am. J. Kidney Dis.: the ofcial
journal of the National Kidney Foundation 38 (5 Suppl. 5) (2001) S3S19.
[25] M. Hewison, F. Burke, K.N. Evans, D.A. Lammas, D.M. Sansom, P. Liu, et al., Extra-
renal 25-hydroxyvitamin D3-1alpha-hydroxylase in human health and disease, J.
Steroid Biochem. Mol. Biol. 103 (3-5) (2007) 316321.
[26] M. van Driel, M. Koedam, C.J. Buurman, M. Roelse, F. Weyts, H. Chiba, et al.,
Evidence that both 1alpha,25-dihydroxyvitamin D3 and 24-hydroxyl ated D3
enhance human osteoblast differentiation and mineralization, J. Cell. Biochem. 99
(3) (2006) 922935.
[27] J. Rojas-Rivera, C. De La Piedra,A. Ramos, A. Ortiz, J. Egido,The expanding spectrum
of biological actions of vitamin D, Nephrol. Dial. Transplant.: ofcial publication of
the European Dialysis and Transplant AssociationEuropean Renal Association 25
(9)(2010)28502865.
[28] K. Kalantar-Zadeh, C.P. Kovesdy, Clinical outcomes with active versus nutritional
vitamin D compounds in chronic kidney disease, Clin. J. Am. Soc. Nephrol.: CJASN
4 (9) (2009) 15291539.
[29] S.U. Nigwekar, I. Bhan, R.Thadhani, Ergocalciferol and cholecalciferol in CKD, Am. J.
Kidney Dis.: the ofcial journal of the National Kidney Foundation 60 (1) (2012)
139156.
Fig. 4. Role of vitamin D in crosstalk between innate and adaptive immunity. Vitamin D plays a role in the crosstalk between innate and adaptive immunity because it has an essential
inuence on macrophages and dendritic cells (DCs). DCs are the key connector between innate and adapti ve immune system s. Local product ion of 1,25-dihydroxyvitamin D
[1,25(OH)
2
D] in paracrine responses also seems to have an effect on monocytes and immature DCs, which express higher levels of vitamin D receptor (VDR) than matureDCs.IFN-γis
an important cytokine for innate and adaptive immunity. T cells express IFN-γthrough the stimulation of DCs. IFN-γincreases the expression of 1α-hydroxylase to augment local
1,25(OH)
2
D production to enhance macrophage phagocytosis.
142 W.-C. Liu et al. / Clinica Chimica Acta 450 (2015) 135144
Author's personal copy
[30] A. Verstuyf, G. Carmeliet, R. Bouillon,C. Mathieu, Vitamin D:a pleiotropic hormone,
Kidney Int. 78 (2) (2010) 140145.
[31] A. Hossein-nezhad, M.F. Holick, Vitamin D for health: a global perspective, Mayo
Clin. Proc. 88 (7) (2013) 720755.
[32] C.L. Lang, M.H. Wang, C.K. Chiang, K.C. Lu, Vitamin D and the immune system from
the nephrologist's viewpoint, ISRN endocrinology 2014 (2014) 105456.
[33] M. Hewison, Vita min D and the immun e system: new pers pectives on an ol d
theme, Endocrinol. Metab. Clin. N. Am. 39 (2) (2010) 365379 (table of contents).
[34] C. Mathieu, M. Jafari, Immunomodulation by 1,25-dihydroxyvitamin D3: therapeutic
implications in hemodialysis and renal transplantation, Clin. Nephrol. 66 (4) (2006)
275283.
[35] K. Casteels, R. Bouillon, M. Waer, C. Mathieu, Immunomodulatory effects of 1,25-
dihydroxyvitamin D3, Curr. Opin. Nephrol. Hypertens. 4 (4) (1995) 313318.
[36] M.F. Holick, Vitamin D, deciency, N. Engl. J. Med. 357 (3) (2007) 266281.
[37] P. Muszkat, M.B. Camar go, L.H. Griz, M. Lazaretti -Castro, Evidence-based non-
skeletal action s of vitamin D, Arq. Bras. Endocrinol. Metabol. 54 (2) (2010)
110117.
[38] A.S. Dusso, M. Tokumoto, Defective renal maintenance of the vitamin D endocrine
system impairs vitamin D renoprotection: a downward spiral in kidney disease,
Kidney Int. 79 (7) (2011) 715729.
[39] D. Zehnder, M.J. Landray, D.C. Wheeler, W. Fraser, L. Blackwell, S. Nuttall, et al.,
Cross-sectional analysis of abnormalities of mineral homeostasis, vitamin D and
parathyroid ho rmone in a cohort of pre-dialysis patients. The c hronic renal
impairment in Birmingham (CRIB) study, Nephr on Clin. Pract. 107 (3) (2007)
c109c116.
[40] A. Tourigny, F. Charbonneau, P. Xing, R. Boukrab, G. Rousseau, R. St-Arnaud, et al.,
CYP24A1 exacerbated activity during diabetes co ntributes to kid ney tubular
apoptosis via caspase-3 incre ased expression and activation, PLoS One 7 (10)
(2012) e48652.
[41] S.Christakos,D.V.Ajibade,P.Dhawan,A.J.Fechner,L.J.Mady,VitaminD:metabolism,
Endocrinol. Metab. Clin. N. Am. 39 (2) (2010) 243253 (table of contents).
[42] T. Larsson, U. Nisbeth,O. Ljunggren, H. Juppner, K.B. Jonsson, Circulating concentration
of FGF-23 increases as renal function declines in patientswith chronic kidney disease,
but does not change in response to variation in phosphate intake in healthy
volunteers, Kidney Int. 64 (6) (2003) 22722279.
[43] D. Prie, G. Friedlander, Reciprocal control of 1,25-dihydroxyvitamin D and FGF23
formation involving the FGF23/Klotho system, J. Am. Soc. Nephrol.: CJASN 5 (9)
(2010) 17171722.
[44] C. Ding, D. Gao, J. Wilding, P. Trayhurn, C. Bing, Vitamin D signalling in adipose
tissue, Br. J. Nutr. 108 (11) (2012) 19151923.
[45] E. Ishimura, Y. Nishizawa, M. Inaba, N. Matsumoto, M. Emoto, T. Kawagishi, et al.,
Serum levels of 1 ,25-dihydrox yvitamin D, 24,25-dihydroxyvitamin D, and
25-hydroxyvitamin D in nondialyzed patients with chronic renal failure, Kidney
Int. 55 (3) (1999) 10191027.
[46] S. Battault, S.J. Whiting, S.L. Peltier, S. Sadrin, G. Gerber, J.M. Maixent, Vitamin D
metabolism, functions and needs: from science to health claims, Eur. J. Nutr. 52
(2) (2013) 429441.
[47] M. Hewison, M.A. Ga cad, J. Lemire, J.S. Adams, Vitamin D as a cytokine and
hematopoetic factor, Rev. Endocr. Metab. Disord. 2 (2) (2001) 217227.
[48] J.S. Adams, M. Hewison, Update in vitamin D, J. Clin. Endocrinol. Metab. 95 (2)
(2010) 471478.
[49] C.A. Peterson, M.E. Heffernan, Serum tumor necrosis factor-alpha concentrations
are negatively correlated with ser um 25(OH)D concentrations in heal thy
women, J. Inamm. 5 (2008) 10.
[50] J.M. Gelfand, B.A. Cree, J. McElroy, J. Oksenberg, R. Gre en, E.M. Mowry, et al .,
Vitamin D in African Americans wi th multiple sclerosis, Neurology 76 (21)
(2011) 18241830.
[51] L.L. Ritterhouse, S.R. Crowe, T.B. Niewold, D.L. Kamen, S.R. Macwana, V.C. Roberts,
et al., Vitamin D deciency is associated with an increased autoimmune response
in healthy individuals and in patients with systemic lupus erythematosus, Ann.
Rheum. Dis. 70 (9) (2011) 15691574.
[52] G. Zhao, E.S. Ford, J. Tsai, C. Li, J.B. Croft, Low concentrations of serum 25-
hydroxyvitamin D associated with increased risk for chronic bronchitis among
US adults, Br. J. Nutr. 107 (9) (2012) 13861392.
[53] G. Schwalfenberg, Not enough vitamin D: health consequences for Canadians,
Canadian family physician Medecin de famille canadien 53 (5) (2007) 841854.
[54] T.T. Wang, F.P. Nestel, V. Bourdeau, Y. Nagai, Q. Wang, J. Liao, et al., Cutting edge:
1,25-dihydroxyvitamin D3 is a direct inducer of antimicrobial peptide gene
expression, J. Immunol. 173 (5) (2004) 29092912.
[55] A.F. Gombart, N. Borregaard, H.P. Koefer, Human cathelicidin antimicr obial
peptide (CAMP) gene is a direct target of the vitamin D receptor and is strongly
up-regulated in myeloid cells by 1, 25-dihydroxy vitamin D3, FASEB J.: ofcial
publication of the Federation of American Societies for Experimental Biology 19
(9) (2005) 10671077.
[56] J.H. White, Vitami n D, metabolism and si gnaling in the immune system, Rev.
Endocr. Metab. Disord. 13 (1) (2012) 2129.
[57] P. Bergman, A.C. Norlin, S.Hansen, R.S. Rekha, B. Agerberth, L. Bjorkhem-Bergman,
et al., Vitamin D3 sup plementation in pa tients with frequent respiratory tract
infections: a randomised and double-blind intervention study, BMJ open 2 (6)
(2012).
[58] H.D. Brightbill, R.L. Modlin, Toll-like receptors: molecular mechanisms of the
mammalian immune response, Immunology 101 (1) (2000) 110.
[59] E. Abe, C. Miyaura, H. Tanaka, Y. Shiina, T. Kuribayashi, S. Suda, et al., 1 alpha,25-
dihydroxyvitamin D3 promotes fusion of mouse alveolar macrophages both by a
direct mechanism and by a spleen cell-mediated indirect mechanism, Proc. Natl.
Acad. Sci. U. S. A. 80 (18) (1983) 55835587.
[60] H. Tanaka, E. Abe, C. Miyaura, Y. Shiina, T. Suda, 1 alpha,25-dihydroxyvitamin D3
induces differe ntiation of human promyelocytic leukemia cells (HL-60 ) into
monocyte-mac rophages, but not into granulocyte s, Biochem. Biophys. Res.
Commun. 117 (1) (1983) 8692.
[61] H.P. Koefer, T. Amatruda, N. Ikekawa , Y. Kobayashi, H.F. DeLuca, Induction of
macrophage differentiation of human normal and leukemic myeloid stem cells
by 1,25-dihydroxyvitamin D3 and its uorinated analogues, Cancer Res. 44 (12
Pt 1) (1984) 56245628.
[62] K. Edfeldt, P.T. Liu, R. Chun, M. Fabri, M. Schen k, M. Wheelwright, et al., T-cell
cytokines diff erentially con trol human monocyte antimicro bial responses by
regulating vitamin D metabolism, Proc. Natl. Acad. Sci. U. S. A. 107 (52) (2010)
2259322598.
[63] D. Andress, No nclassical aspects of differential vitamin D receptor activation:
implications for survival in patients with chronic kidney disease, Drugs 67 (14)
(2007) 19992012.
[64] B. Ramanathan, E.G. Davis, C.R. Ross, F. Blecha, Cathelicidins: microbicidal activity,
mechanisms of action, and roles in innate immunity, Mic robes and infect ion/
Institut Pasteur 4 (3) (2002) 361372.
[65] M. Hewison, Antibacterial effects of vitamin D, Nat. Rev. Endocrinol. 7 (6) (2011)
337345.
[66] J.A. Beard, A. Bearden, R. Striker, Vitamin D and the anti-viral state, J. Clin. Virol.:
the ofcial publication of the Pan American Society for Clinical Virology 50 (3)
(2011) 194200.
[67] L. Jeng, A.V. Yamshchikov, S.E. Judd, H.M. Blumberg, G.S. Martin, T.R. Ziegler, et al.,
Alterations in vitamin D status and anti-microbial peptide levels in patients in the
intensive care unit with sepsis, J. Transl. Med. 7 (2009) 28.
[68] K. Stoffels, L. Overbergh, R. Bouillon, C. Mathieu, Immune regulation of 1alpha-
hydroxylase in murine peritoneal macrophages: unravelling the IFNgamma
pathway, J. Steroid Biochem. Mol. Biol. 103 (3-5) (2007) 567571.
[69] L. Esteban, M. Vidal, A. Dusso, 1a lpha-Hydroxylase transac tivation by gamma-
interferon in murine macrophages requires enhanced C/EBPbeta expression and
activation, J. Steroid Biochem. Mol. Biol. 89-90 (1-5) (2004) 131137.
[70] B. Sakem, C. Nock, Z. Stanga, P. Medina, U.E. Nydegger, M. Ri sch, et al., Serum
concentrations of 25-hydroxyvitamin D and immunoglobulins in an older Swiss
cohort: results of the Senior Labor Study, BMC Med. 11 (2013) 176.
[71] A.F. Gombart, I. Bhan, N. Borregaard, H. Tamez, C.A. Camargo Jr., H.P. Koefer,etal.,
Low plasma level of cathelicidin antimicrobial peptide (hCAP18) predicts
increased infectious disease mortality in patients undergoing hemodialysis, Clin.
Infect. Dis.: an ofcial publication of the Infectious Diseases Society of America
48 (4) (2009) 418424.
[72] S. Yim, P. Dhawan, C. Ragunath, S. Christakos, G. Diamond, Induction of cathelicidin
in normal and CF bronchial epithelial cells by 1,25-dihydroxyvitamin D(3), J. Cyst.
Fibros.: ofcial journal of the European Cystic Fibrosis Society 6 (6) (2007) 403410.
[73] D.A. O'Neil, E.M. Porter, D. Elewaut, G.M. Anderson, L. Eckmann, T. Ganz, et al.,
Expression and re gulation of the huma n beta-defensins hBD-1 and hBD-2 in
intestinal epithelium, J. Immunol. 163 (12) (1999) 67186724.
[74] W. Strober, P.J. Murray, A. Kitani, T. Watanabe, Signalling pathways and molecular
interactions of NOD1 and NOD2, Nat. Rev. Immunol. 6 (1) (2006) 920.
[75] P.T. Liu, M. Sch enk, V.P. Walker, P. W. Dempsey, M. Kan chanapoomi, M.
Wheelwright, et al ., Convergence of IL-1beta and VDR acti vation pathway s in
human TLR2/1-induced antimicrobial responses, PLoS One 4 (6) (2009) e5810.
[76] T.T. Wang, B. Dabbas, D. Laperriere, A.J. Bitton, H. Soualhine, L.E. Tavera-Mendoza,
et al., Direct and indirect induction by 1,25-dihydroxyvitamin D3 of the NOD2/
CARD15-defensin beta2 innate immune pathway defective in Crohn disease, J.
Biol. Chem. 285 (4) (2010) 22272231.
[77] M. Hewison, Vitamin D, and innate and adaptive immunity, Vitam. Horm. 86
(2011) 2362.
[78] J.M. Yuk, D.M. Shin, H.M. Lee, C.S. Yang, H.S.Jin, K.K. Kim, et al., Vitamin D3 induces
autophagy in human monocytes/macrophages via cathelicidin, CellHost Microbe 6
(3)(2009)231243.
[79] B. Levine, N. Mizushima, H.W. Virgin, Autophagy in immunity and inammation,
Nature 469 (7330) (2011) 323335.
[80] D.M. Provvedini, C.D. Tsoukas, L.J. Deftos, S.C. Manolagas, 1,25-dihydroxyvitamin
D3 receptors in human leukocytes, Science 221 (4616) (1983) 11811183.
[81] A.K. Bhalla, E.P. Amento, T.L. Clemens, M.F. Holick, S.M. Krane, Specichigh-afnity
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. 57 (6) (1983) 13081310.
[82] D.M. Provvedi ni, C.D. Tsoukas, L.J. Deftos, S.C. Manolag as, 1 alpha,25-
Dihydroxyvitamin D3-binding macromolecules in human B lymphocytes: effects
on immunoglobulin production, J. Immunol. 136 (8) (1986) 27342740.
[83] M.D. Grifn, W.H. Lutz, V.A. Phan, L.A. Bachman, D.J. McKean, R. Kumar, Potent in-
hibition of dendritic cell differentiat ion and maturation b y vitamin D analogs,
Biochem. Biophys. Res. Commun. 270 (3) (2000) 701708.
[84] M.D. Grifn, W. Lutz, V.A.Phan, L.A. Bachman,D.J. McKean, R. Kumar, Dendritic cell
modulation by 1alp ha,25 dihydroxy vitamin D3 and its analogs: a vitamin D
receptor-dependent pathway th at promotes a persistent state o f immaturity
in vitro and in vivo, Proc. Natl. Acad. Sci. U. S. A. 98 (12) (2001) 68006805.
[85] G. Penna, L. Adori ni, 1 Alpha,25-dihydroxyvitamin D3 inhibits differentiation,
maturation, acti vation, and surv ival of dendriti c cells leading to impaired
alloreactive T cell activation, J. Immunol. 164 (5) (2000) 24052411.
[86] L. Piemonti, P. Monti, M.Sironi, P. Fraticelli, B.E. Leone, E. Dal Cin, et al., Vitamin D3
affects differentiation, matu ration, and function of human mo nocyte-derive d
dendritic cells, J. Immunol. 164 (9) (2000) 44434451.
[87] X. Dong, T. Craig, N. Xing, L.A. Bachman, C.V. Paya, F. Weih, et al., Direct transcrip-
tional regulati on of RelB by 1alpha,25-dihydroxyvitamin D3 an d its analogs:
143W.-C. Liu et al. / Clinica Chimica Acta 450 (2015) 135144
Author's personal copy
physiologic and therapeutic implications for dendritic cell function, J. Biol. Chem.
278 (49) (2003) 4937849385.
[88] X. Dong, W. Lutz, T.M. Schroeder, L.A. Bachman, J.J. Westendorf, R. Kumar, et al.,
Regulation of relB in dendritic cells by means of modulated association of vitamin D
receptor and histone deacetylase 3 with the promoter, Proc. Natl. Acad. Sci. U. S. A.
102 (44) (2005) 1600716012.
[89] E. van Etten, C. Mathieu, Immunoregulation by 1,25-dihydroxyvitamin D3: basic
concepts, J. Steroid Biochem. Mol. Biol. 97 (1-2) (2005) 93101.
[90] E. Seibert, N.W. Levin, M.K. Kuhlmann, Immunomodulating effects of vitamin D
analogs in hemo dialysis patien ts, Hemodialysis internatio nal International
Symposium on Home Hemodialysis 9 (Suppl. 1) (2005) S25S29.
[91] M. Hewison, L. Freeman, S.V. Hughes, K.N. Evans, R. Bland, A.G. Eliopoulos, et al.,
Differential regulation of vitamin D receptor and its ligand in human monocyte-
derived dendritic cells, J. Immunol. 170 (11) (2003) 53825390.
[92] D. D'Ambrosio, M. Cippitelli, M.G. Cocciolo, D. Mazzeo, P. Di Lucia, R. Lang, et al.,
Inhibition of IL- 12 production by 1,25- dihydroxyvitamin D3. Involvem ent of
NF-kappaB downregulation in transcriptional repression of the p40 gene, J. Clin.
Invest. 101 (1) (1998) 252262.
[93] J. Fritsche, K. Mo ndal, A. Ehrnsperger, R. Andrees en, M. Kreutz, Regulation of
25-hydroxyvitamin D3-1 alpha -hydroxylase and production of 1 alpha,25-
dihydroxyvitamin D3 by human dendritic cells, Blood 102 (9) (2003) 33143316.
[94] M. Hewison, D. Zehnder, R. Chakraverty, J.S. Adams, Vitamin D and barrier
function: a novel role for extra-renal 1 alpha-hydroxylase, Mol. Cell. Endocrinol.
215 (1-2) (2004) 3138.
[95] J.M. Lemire, J.S. Adams, R. Sakai, S.C. Jordan, 1 alpha,25-dihydroxyvitamin D3
suppresses proliferation and immunoglobulin produ ction by normal human
peripheral blood mononuclear cells, J. Clin. Invest. 74 (2) (1984) 657661.
[96] A. Boonstra, F.J. Barrat, C. Crain, V.L. Heath, H.F. Savelkoul, A. O'Garra, 1alpha,25-
Dihydroxyvitamin d3 has a direct effect on naive CD4(+) T cells to enhance the
development of Th2 cells, J. Immunol. 167 (9) (2001) 49744980.
[97] K. Takahashi, H.Horiuchi, T. Ohta, K. Komoriya, H. Ohmori, T.Kamimura, 1 alpha,25-
dihydroxyvitamin D3 suppresses interleukin-1beta-induced interleukin-8 produc-
tion in human whole blood: an involvement of erythrocytes in the inhibition,
Immunopharmacol. Immunotoxicol. 24 (1) (2002) 115.
[98] H. Xu, A. Soruri, R.K. Gieseler, J.H. Peters, 1,25-Dihydroxyvitamin D3 exerts oppos-
ing effects to IL-4 on MHC class- II antigen expr ession, accessory activity, a nd
phagocytosis of human monocytes, Scand. J. Immunol. 38 (6) (1993) 535540.
[99] M. Hewison, An update on vitamin D and human immunity, Clin. Endocrinol. 76
(3) (2012) 315325.
[100] I.I. Ivanov, B.S. McKenzie, L. Zhou, C.E. Tadokoro, A. Lepelley, J.J. Lafaille, et al., The
orphan nuclear receptor RORgammat directs the differen tiation program of
proinammatory IL-17+ T helper cells, Cell 126 (6) (2006) 11211133.
[101] G. Penna, S. Amuchastegui, C. Cossetti, F. Aquilano, R. Mariani, F. Sanvito, et al.,
Treatment of experimental autoimmune prostatitis in nonobese diabetic mice by
the vitaminD receptoragonist elocalcitol,J. Immunol. 177 (12) (2006)85048511.
[102] D. Bikle, Nonclassic actions of vitamin D, J. Clin. Endocrinol. Metab. 94 (1) (2009)
2634.
[103] C.L. Lang, M.H. Wang, K.Y. Hung, C.K. Chiang, K.C. Lu, Altered molecular repertoire
of immune system by renal dysfunction in the elderly: is prediction and targeted
prevention in the horizon? EPMA J. 4 (1) (2013) 17.
[104] C.L. Lang, M.H. Wang, K.Y. Hung, S.H. H su, C.K. Chiang, K .C. Lu, Correlat ion of
interleukin-17-producing effector memory T cells and CD4
+
CD25
+
Foxp3 regula-
tory T cells with the phosphate level s in chronic hemodi alysis patients ,
TheScienticWorldJOURNAL 2014 (2014) 593170.
[105] S.Sakaguchi, M.Ono, R. Setoguchi, H. Yagi,S. Hori, Z. Fehervari, et al.,Foxp3
+
CD25
+
CD4
+
natural regulatory T cells in dominant self-tolerance andautoimmunedisease,
Immunol. Rev. 212 (2006) 827.
[106] J.S.Adams, M. Hewison, Unexpected actions of vitaminD: new perspectives on the
regulation of innate and adaptive immunity, Nat. Clin. Pract. Endocrinol. Metab. 4
(2) (2008) 8090.
[107] G. Penna, S. Amuchastegui, N. Giarratana, K.C. Daniel, M. Vulcano, S. Sozzani, et al.,
1,25-DihydroxyvitaminD3 selectively modulates tolerogenic properties in myeloid
but not plasmacytoid dendritic cells, J. Immunol. 178 (1) (2007) 145153.
[108] S. Sakaguchi, T. Yamaguchi, T. Nomura, M. Ono, Regulatory T cells and immune
tolerance, Cell 133 (5) (2008) 775787.
[109] B. Prietl, G. Treiber , T.R. Pieber, K. Amr ein, Vitamin D and imm une function,
Nutrients 5 (7) (2013) 25022521.
[110] B. Prietl, S. Pilz, M. Wolf, A. Tomaschitz,B. Obermayer-Pietsch, W. Graninger, et al.,
Vitamin D supplementation and regulatory T cells in apparently healthy subjects:
vitamin D treatment for autoimmune diseases? Isr. Med. Assoc. J.: IMAJ 12 (3)
(2010) 136139.
[111] M.R. Ardalan, H. Maljaei, M.M. Shoja, A.R. Piri, H.T. Khosroshahi, H. Noshad, et al.,
Calcitriol started in the donor, expands the population of CD4
+
CD25
+
T cells in
renal transplant recipients, Transplant. Proc. 39 (4) (2007) 951953.
[112] L. Overbergh, B. Decallonne, M. Waer, O. Rutgeerts, D. Valckx, K.M. Casteels, et al.,
1alpha,25-dihydroxyv itamin D3 induces an autoantigen-specic T-helper 1/T-
helper 2 immune shift in NOD mice immunized withGAD65 (p524543), Diabetes
49 (8) (2000) 13011307.
[113] S. Chen, G.P. Sims, X.X. Chen, Y.Y. Gu, S. Chen, P.E. Lipsky, Modulatory effects of
1,25-dihydroxyvitamin D3 on human B cell differentiation, J. Immunol. 179 (3)
(2007) 16341647.
[114] M.T. Abreu, V. Kantorovich, E.A. Vasiliauskas, U. Gruntmanis, R. Matuk, K. Daigle,
et al., Measurement of vitamin D levels in inammatory bowel disease patients
reveals a subset of Crohn's disease patients with elevated 1,25-dihydroxyvitamin D
and low bone mineral density, Gut 53 (8) (2004) 11291136.
[115] C.C. Sung, M.T. Liao, K. C. Lu, C.C. Wu, Role of vitamin D in insulin resistance, J.
Biomed. Biotechnol. 2012 (2012) 634195.
[116] K.C. Hung, C.C. Wu, H.S. Chen, W.Y. Ma, C.F. Tseng, L.K. Yang, et al., Serum IL-6,
albumin and co-morbidities are closely correlated with symptoms of depression
in patients on maintenance haemodialysis, Nephrol . Dial. Transplant.: ofcial
publication of the European Dialysis and Tr ansplant Assoc iation European
Renal Association 26 (2) (2011) 658664.
[117] T. Eleftheriadis, G. Antoniadi, V. Liakopoulos, C. Kartsios, I. Stefanidis, Disturbances
of acquired immun ity in hemodialysis patients, Semin. Dial. 20 (5) (2 007)
440451.
[118] A.J. Matas, R.L. Simmons, C.M. Kjellstrand, T.J. Buselmeier, J.S. Najarian, Increased
incidence of malignancy during chronic renal failure, Lancet 1 (7912 ) (1975)
883886.
[119] B. Kreft, M. Klouche, R. Kreft, H. Kirchner, K. Sack, Low efciency of active immuni-
zation against diphtheria in ch ronic hemodia lysis patients , Kidney Int. 52 (1)
(1997) 212216.
[120] R. Schindler, S. Linnenweber, M. Schulze, M. Oppermann,C.A. Dinarello, S. Shaldon,
et al., Gene expression of interleukin-1 beta duringhemodialysis, KidneyInt. 43 (3)
(1993) 712721.
[121] M. Girndt, H. Kohler, E. Schiedhelm-Weick, J.F. Schlaak, Meyer zum Buschenfelde
KH, Fleischer B. Production of interleukin-6, tum or necrosis fac tor alpha and
interleukin-1 0 in vitro correlat es with the clinical immune defect in chronic
hemodialysis patients, Kidney Int. 47 (2) (1995) 559565.
[122] M. Rao, C. Wong, P. Kanetsky, M. Girndt, P. Stenvinkel, M. Reilly, et al., Cytokine
gene polymorphism and progression of renal and cardiovascular diseases, Kidney
Int. 72 (5) (2007) 549556.
[123] D.S. Raj, M.A. Boivin, E.A. Dominic, A. Boyd, P.K. Roy, T. Rihani,et al., Haemodialysis
induces mitochondrial dysfunction and apoptosis, Eur. J. Clin. Investig. 37 (12)
(2007) 971977.
[124] C.E. Stevens, H.J. Alter, P.E. Taylor, E.A. Zang, E.J. Harley, W. Szmuness, Hepatitis B
vaccine in patients receiving hemodialysis. Immunogenicity and efcacy, N. Engl.
J. Med. 311 (8) (1984) 496501.
[125] E. Zitt, H. Sprenger-Mahr, F. Knoll, U. Ney er, K. Lhotta, Vitamin D deciency is
associated with poor respons e to active hepati tis B immunisation in patients
with chronic kidney disease, Vaccine 30 (5) (2012) 931935.
[126] M. Girndt, M. Sester, U. Sester, H. Kaul, H. Kohler, Defective expression of B7-2
(CD86) on monocytes of dialysis patients corre lates to the uremia-associated
immune defect, Kidney Int. 59 (4) (2001) 13821389.
[127] S. Agrawal, P. Gollapudi, R. Elahimehr, M.V. Pahl, N.D. Vaziri, Effects of end-stage
renal disease and h aemodialysis on dendritic cell sub sets and basal and
LPS-stimulated cytokine production, Nephrol. Dial. Transplant.: ofcial publication
of the European Dialysis and Transplant Association European Renal Association
25 (3) (2010) 737746.
[128] W.H. Lim, S. Kireta, E. Leedham, G.R. Russ, P.T. Coates, Uremia impairs monocyte
and monocyte-derived dendritic cell function in hemodialysis patients, Kidney
Int. 72 (9) (2007) 11381148.
[129] K.A. Lisowska, A. Debska-Slizien, A. Jasiulewicz, Z. Heleniak, E. Bryl, J.M. Witkowski,
Hemodialysis affects phenotype and proliferation of CD4-positive T lymphocytes, J.
Clin. Immunol. 32 (1) (2012) 189200.
[130] B. Descamps-Latscha, A. Herbelin, A.T. Nguyen, P. Roux-Lombard, J. Zingraff, A.
Moynot, et al., Balance between IL-1 beta, TNF-alpha, and their specic inhibitors
in chronic renal failure and maintenance dialysis. Relationships with activation
markers of T cells, B cells, and monocytes, J. Immunol. 154 (2) (1995) 882892.
[131] P. Stenvinkel, M. Ketteler, R.J. Johnson, B. Lindholm, R. Pecoits-Filho, M.Riella, et al.,
IL-10, IL-6, and TN F-alpha: centr al factors in the altered cytokine network of
uremiathe good, the bad, and the ugly, Kidney Int. 67 (4) (2005) 12161233.
[132] M. Cutolo, C. Pizzorn i, A. Sulli, Vitamin D endocrine syste m involvement in
autoimmune rheumatic diseases, Autoimmun. Rev. 11 (2) (2011) 8487.
[133] J.C. Guery, L. Adorini, Dendritic cells are the most efcient in presenting
endogenous natu rally processed self-epitopes to class II-restricte d T cells, J.
Immunol. 154 (2) (1995) 536544.
[134] R.H. Schwartz, A cell culture model for T lymphocyte clonal anergy, Science 248
(4961) (1990) 13491356.
[135] D.D. Bikle, Vitamin D, regulation of i mmune function, Vitam. Horm. 86 (2011)
121.
[136] J.R. Schoenborn, C.B. Wilson, Regulation of interferon-gamma during innate and
adaptive immune responses, Adv. Immunol. 96 (2007) 41101.
[137] R. Josien, B.R. Wong, H.L. Li, R.M. Steinman, Y. Choi, TRANCE, a TNF family member,
is differentially expressed on T cell subsets and induces cytokine production in
dendritic cells, J. Immunol. 162 (5) (1999) 25622568.
144 W.-C. Liu et al. / Clinica Chimica Acta 450 (2015) 135144
... The immunomodulatory effect of Vitamin D, and especially its action on T-cells, with the shift toward a less inflammatory and a more tolerogenic phenotype, could be responsible for its potential counteraction of chronic allograft dysfunction, thus enhancing graft survival [107][108][109][110][111]. Despite these considerations, data on the clinical effectiveness of Vitamin D supplementation for prolonging graft survival are controversial; furthermore, many of the available evidences come from observational studies rather than randomized controlled trials [40,[112][113][114][115]. ...
Preprint
Full-text available
Chronic kidney disease (CKD) and cardiovascular disease (CVD) are highly prevalent conditions, each significantly contributing to the global burden of morbidity and mortality. CVD and CKD share a great number of common risk factor, such as hypertension, diabetes, obesity, and smoking among others. Their relationship extends beyond these factors, encompassing intricate interplay between the two systems. Within this complex network of pathophysiological processes, vitamin D has emerged as a potential linchpin, exerting influence over diverse physiological pathways implicated in both CKD and CVD. In recent years, scientific exploration has unveiled a close connection between these two prevalent conditions and vitamin D, a crucial hormone traditionally recognized for its role in bone health. This article aims to provide an extensive review of vitamin D's multifaceted and expanding actions concerning its involvement in CKD and CVD.
... Additionally, VitD increases the production of T helper (Th)2 chemokines and improves the efficiency of Treg lymphocytes, while inhibiting the secretion of Th1 and Th17 chemokines. VitD plays a crucial role in maintaining normal immune function and facilitating communication between the innate and adaptive immune systems [27]. In the context of inflammatory bowel disease, VitD can also promotes macrophage M2 polarization [28], downregulate the activity of dendritic cells, induce tolerogenic T cell differentiation and function instead of pro-inflammatory responses, and increase the levels of anti-inflammatory chemokines through the activation of VDR, underscoring its significance [29]. ...
Article
Full-text available
Background Activation of VDR pathway was a promising anti-tumor therapy strategy. However, numerous clinical studies have demonstrated the effect of activating VDR is limited, which indicates that VDR plays a complex role in vivos. Methods We analyzed the TCGA database to examine the association between VDR expression and immune cell infiltration in pancreatic adenocarcinoma (PAAD). Western blot, ELISA, ChIP, and dual-luciferase reporter assays were performed to determine the mechanism of VDR regulating CCL20. Migration assay and immunofluorescence were used to investigate the role of CCL20 in M2 macrophage polarization and recruitment. We employed multiplexed immunohistochemical staining and mouse models to validate the correlation of VDR on macrophages infiltration in PAAD. Flow cytometry analysis of M2/M1 ratio in subcutaneous graft tumors. Results VDR is extensively expressed in PAAD, and patients with elevated VDR levels exhibited a significantly reduced overall survival. VDR expression in PAAD tissues was associated with increased M2 macrophages infiltration. PAAD cells overexpressing VDR promote macrophages polarization towards M2 phenotype and recruitment in vitro and vivo. Mechanistically, VDR binds to the CCL20 promoter and up-regulates its transcription. The effects of polarization and recruitment on macrophages can be rescued by blocking CCL20. Finally, the relationship between VDR and M2 macrophages infiltration was evaluated using clinical cohort and subcutaneous graft tumors. A positive correlation was demonstrated between VDR/CCL20/CD163 in PAAD tissues and mouse models. Conclusion High expression of VDR in PAAD promotes M2 macrophage polarization and recruitment through the secretion of CCL20, which activates tumor progression. This finding suggests that the combination of anti-macrophage therapy may improve the efficacy of VDR activation therapy in PAAD.
... Subgroup analyses were performed regardless of the statistical significance of interaction. We further performed three sensitivity analyses (excluding those whose eGFR < 60 mL/min/ 1.73 m 2 [30], whose daily calorie intake <1,000 kcal [31], and who were recruited from Xin Hua Hospital) to test the robustness of main analysis. A P-values < 0.05 was considered significant, and the analysis of the data was carried out using SAS software (SAS Institute, Inc., Cary, NC, USA). ...
Article
Full-text available
Objective The association between vitamin D status and inflammation remains unclear in hospitalized patients. Materials and Methods We performed the current study based on real-world data from two teaching hospitals. Serum level of vitamin D (assessed by 25-hydroxyvitamin D) was evaluated within 2 days after admission. All the patients were further classified into three groups: deficiency (<12 ng/mL), insufficiency (12–20 ng/mL), and adequate (≥20 ng/mL). White blood cell (WBC) count, serum level of C-reactive protein (CRP), and procalcitonin were also measured and used to evaluate inflammation. Other potential covariates were abstracted from medical records. Charlson comorbidity index (CCI) was calculated to assess the severity of disease. Results A total number of 35,528 hospitalized adult patients (21,171 men and 14,357 women) were included. The average age and BMI were 57.5 ± 16.2 years and 23.4 ± 3.7 kg/m², respectively, while medium vitamin D level was 16.1 ng/mL (interquartile range: 11.4 ng/mL, 21.6 ng/mL) and median CCI was one point (interquartile range: 0 point, two points). The prevalence of deficiency and insufficiency was 28.0% and 40.5%. Multivariate linear regression model showed that serum level of vitamin D was significantly associated with WBC and CRP but not associated with procalcitonin. Each standard deviation (≈7.4 ng/mL) increase in vitamin D was associated with a decrease in WBC by 0.13 × 10⁹/mL (95% CI: 0.2 × 10⁹/mL, 0.06 × 10⁹/mL) and 0.62 mg/L (95% CI: 0.88 mg/L, 0.37 mg/L) for CRP. Subgroup analysis and sensitivity analysis (excluding those whose eGFR <60 ml/min/1.73 m², those whose daily calorie intake <1,000 kcal, and those who were recruited from Xin Hua hospital) generated similar results. Conclusions The deficiency and insufficiency of vitamin D in the hospitalized adult patients was very common. However, the results should be interpreted with caution for limited representation of the whole inpatients. Low level of vitamin D was associated with inflammatory biomarkers, which provide the evidences to early intervention for lower the risk of infection.
... Thus, vitamin D may enhance the innate immune response against the virus and may dampen the hyperinflammatory reaction against the COVID-19 virus [59]. Vitamin D enhances the innate immune response against the SARS-CoV-2 virus by activating toll-like receptor 2 and via peptide synthesis, which acts to combat infectious invasions [62]. Vitamin D decreases proinflammatory cytokine release via CD4+ lymphocytes, thus inhibiting the development of a cytokine storm [63]. ...
Article
Full-text available
The SARS-CoV-2 virus may cause severe infection, which is associated with diverse clinical manifestations. Vitamin D has immunomodulating properties and may enhance the body’s defense system against invading pathogenic organisms. The aim was to assess 25(OH)D3 levels in patients hospitalized for severe infection from the SARS-CoV-2 virus and explore the relationship between 25(OH)D3 and outcomes. In a group of 88 patients hospitalized for severe infection from the SARS-CoV-2 virus and a control group matched for age and sex, the levels of 25(OH)D3 were analyzed. Levels of 25(OH)D3 were 17.36 ± 8.80 ng/mL (mean ± SD) compared with 24.34 ± 10.34 ng/mL in patients with severe SARS-CoV-2 infection and the control group, respectively, p < 0.001 (Student’s t-test). 25(OH)D3 levels were significantly related to outcomes, i.e., survival as opposed to non-survival, as more patients with 25(OH)D3 deficiency (0–10 ng/mL) and insufficiency (10–20 ng/mL) had a fatal outcome as compared with those with vitamin D sufficiency (p < 0.001, chi-square test, p < 0.001, Fisher’s exact test). Levels of 25(OH)D3 were inversely related to C-reactive protein (CRP), ferritin, d-dimer, and fibrinogen levels (p < 0.001, linear regression analysis, beta coefficient of variation, −0.176, −0.160, −0.178, and −0.158, respectively). Vitamin D deficiency observed in severe SARS-CoV-2 infection was related to disease outcomes.
... Thus, vitamin D may enhance the innate immune response against the virus and may dampen the hyperinflammatory reaction against the COVID-19 virus [51]. Vitamin D enhances the innate immune response against the SARS-CoV-2 virus by activating toll-like receptor 2 and by peptide synthesis, which act to combat infectious invasions [54]. Vitamin D attenuates the release of proinflammatory cytokines by CD4+ lymphocytes thus inhibiting the development of a cytokine storm [55]. ...
Preprint
Full-text available
Severe infection from the SARS-CoV-2 virus is associated with various clinical findings, including hematological manifestations. Thrombotic events or a tendency to develop thrombotic events also characterize severe COVID-19 disease and may lead to death. Vitamin D is known to have immunomodulating properties and to enhance the body defense system against invading pathogens and to have immunostimulatory properties as far as the innate immune response is concerned. The aim was to measure 25(OH)D3 levels in patients hospitalized for severe COVID-19 infection, to explore the relationship between 25(OH)D3 and outcome and to investigate the relationship between 25(OH)D3 levels, CRP, ferritin, d-dimers and fibrinogen levels in this cohort. In a cohort of 88 patients hospitalized for severe infection from the SARS-CoV-2 virus and a control group matched for age and sex the levels of 25(OH)D3 were measured. In the same cohort CRP, ferritin, d-dimer and fibrinogen levels were also analyzed. Levels of 25(OH)D3 were 17.36±8.80 ng/ml (mean±SD) as compared with 24.34±10.34 ng/ml, in patients with severe SARS-CoV-2 infection and the control group, respectively, p<0.001 (Student’s t test). The levels of 25(OH)D3 were found to be significantly related to outcome, i.e. survival as opposed to non-survival, as more patients with 25(OH)D3 deficiency (0-10 ng/ml) and insufficiency (10-20 ng/ml) had a fatal outcome as compared with those with vitamin D sufficiency, p<0.001, chi-square test, p<0.001, Fischer’s exact test. Levels of 25(OH)D3 were inversely related to CRP, ferritin, d-dimer and fibrinogen levels, p<0.001, linear regression analysis, beta coefficient of variation, -0.176, -0,160, -0.178, -0.158, respectively. Vitamin D deficiency observed in severe SARS-CoV-2 infection was related to the disease outcome. Increased ferritin levels, and increased thrombotic markers, namely d-dimer and fibrinogen levels were observed in the patients and their concentration was also inversely related to vitamin D. We showed that vitamin D levels were low in hospitalized patients with severe SARS-CoV-2 infection and also inversely related to CRP, ferritin, d-dimer and fibrinogen concentration. It is proposed that vitamin D levels may be an index of severity in the context of SARS-CoV-2 infection.
... Th17 differentiation is induced by TGF-β, IL-1β, and IL-6 [113], while AhR contributes to Th17 cell differentiation by controlling Stat1 activation [114]. CKD patients experience oxidative stress, cardiovascular disease, and endothelial cell inflammation due to the abnormal activation of the immune system cascade through IS-activated AhR signaling [112,115]. ...
Article
Full-text available
Patients with chronic kidney disease (CKD) often experience a high accumulation of protein-bound uremic toxins (PBUTs), specifically indoxyl sulfate (IS) and p-cresyl sulfate (pCS). In the early stages of CKD, the buildup of PBUTs inhibits bone and muscle function. As CKD progresses, elevated PBUT levels further hinder bone turnover and exacerbate muscle wasting. In the late stage of CKD, hyperparathyroidism worsens PBUT-induced muscle damage but can improve low bone turnover. PBUTs play a significant role in reducing both the quantity and quality of bone by affecting osteoblast and osteoclast lineage. IS, in particular, interferes with osteoblastogenesis by activating aryl hydrocarbon receptor (AhR) signaling, which reduces the expression of Runx2 and impedes osteoblast differentiation. High PBUT levels can also reduce calcitriol production, increase the expression of Wnt antagonists (SOST, DKK1), and decrease klotho expression, all of which contribute to low bone turnover disorders. Furthermore, PBUT accumulation leads to continuous muscle protein breakdown through the excessive production of reactive oxygen species (ROS) and inflammatory cytokines. Interactions between muscles and bones, mediated by various factors released from individual tissues, play a crucial role in the mutual modulation of bone and muscle in CKD. Exercise and nutritional therapy have the potential to yield favorable outcomes. Understanding the underlying mechanisms of bone and muscle loss in CKD can aid in developing new therapies for musculoskeletal diseases, particularly those related to bone loss and muscle wasting.
Article
Full-text available
Chronic kidney disease (CKD) and cardiovascular disease (CVD) are highly prevalent conditions, each significantly contributing to the global burden of morbidity and mortality. CVD and CKD share a great number of common risk factors, such as hypertension, diabetes, obesity, and smoking, among others. Their relationship extends beyond these factors, encompassing intricate interplay between the two systems. Within this complex network of pathophysiological processes, vitamin D has emerged as a potential linchpin, exerting influence over diverse physiological pathways implicated in both CKD and CVD. In recent years, scientific exploration has unveiled a close connection between these two prevalent conditions and vitamin D, a crucial hormone traditionally recognized for its role in bone health. This article aims to provide an extensive review of vitamin D’s multifaceted and expanding actions concerning its involvement in CKD and CVD.
Article
Full-text available
Nutrition management is fundamental for children with chronic kidney disease (CKD). Fluid balance and low-protein and low-sodium diets are the more stressed fields from a nutritional point of view. At the same time, the role of micronutrients is often underestimated. Starting from the causes that could lead to potential micronutrient deficiencies in these patients, this review considers all micronutrients that could be administered in CKD to improve the prognosis of this disease.
Article
Full-text available
Vitamin D and its analogues are widely used as treatments by clinical nephrologists, especially when treating chronic kidney disease (CKD) patients with secondary hyperparathyroidism. As CKD progresses, the ability to compensate for elevations in parathyroid hormone (PTH) and fibroblast growth factor-23 and for decreases in 1,25(OH)2D3 becomes inadequate, which results in hyperphosphatemia, abnormal bone disorders, and extra-skeletal calcification. In addition to its calciotropic effect on the regulation of calcium, phosphate, and parathyroid hormone, vitamin D has many other noncalciotropic effects, including controlling cell differentiation/proliferation and having immunomodulatory effects. There are several immune dysregulations that can be noted when renal function declines. Physicians need to know well both the classical and nonclassical functions of vitamin D. This review is an analysis from the nephrologist's viewpoint and focuses on the relationship between the vitamin D and the immune system, together with vitamin's clinical use to treat kidney diseases.
Article
Full-text available
Background and objectives: Hyperparathyroidism and hyperphosphatemia contribute to the inflammatory effects in chronic hemodialysis (HD) patients. Interleukin-17-producing CD4+ effector memory T (Th17) cells and CD4+CD25+Foxp3 regulatory T (Treg) cells both play critical roles in immune activation and inflammation. We investigated the relationship between the Treg and Th17 cells and the phosphate level in chronic HD patients. Methods: 105 patients aged ≥35 years on chronic HD over 3 months were enrolled. The peripheral blood mononuclear cells were collected, cultured, and stimulated by phytohemagglutinin-L, phorbol myristate acetate, and ionomycin at different time points for T cell differentiation. Results: The T cell differentiation was as follows: Th17 cells (mean±standard deviation (SD): 25.61%±10.2%) and Treg cells (8.45%±4.3%). The Th17 cell differentiation was positively correlated with the phosphate and albumin levels and negatively correlated with age. The Treg cell differentiation was negatively correlated with albumin level and age. In the nondiabetes group (n=53), the Th17 cell differentiation was predominantly correlated with the phosphate and iPTH (intact parathyroid hormone) levels as well as the dialysis vintage. Conclusion: Higher phosphate and iPTH levels and longer dialysis duration may increase Th17 cell differentiation, especially in the nondiabetic chronic HD patients.
Article
Full-text available
Vitamin D and the components of humoral immunity play important roles in human health. Older people have lower 25-hydroxyvitamin D (25(OH)D) serum levels than younger adults. We aimed to determine the levels of 25(OH)D serum concentrations in healthy senior citizens and to study their relationship to the levels of components of humoral immunity. A total of 1,470 healthy Swiss men and women, 60 years or older, were recruited for this study. A total of 179 subjects dropped out of the study because of elevated serum concentrations of C-reactive protein. Fasting blood sera were analyzed for 25(OH)D with the high-performance liquid chromatography (HPLC) and for parathyroid hormone (PTH), immunoglobulins and complement C4 and C3 concentrations with immunoassays. The percentage of participants in each of the four 25(OH)D deficiency groups - severely deficient (<10 ng/ml), deficient (10 to 20), insufficient (21 to 30 ng/ml) and normal (>31 ng/ml) - were statistically compared. The relationship of the major components of the humoral system and age with 25(OH)D levels was also assessed. About 66% of the subjects had insufficient levels of 25(OH)D. Normal levels of 25(OH)D were found in 26.1% of the subjects of which 21% were males and 30.5% were females (total study population). Severely deficient levels of 25(OH)D were found in 7.98% of the total study population. Low levels of 25(OH)D were positively associated with IgG2 (P < 0.01) and with C4 (P < 0.02), yet were inversely related to levels of IgG1 and IgA (P < 0.05) and C3 (P < 0.01). Serum levels of total IgA, IgG, IgG2 and IgG4 peaked together with 25(OH)D during late summer. Approximately two-thirds of the healthy, older Swiss population presented with Vitamin D insufficiency. The incremental shift in IgA and C3 levels might not necessarily reflect a deranged humoral immune defense; however, given the high prevalence of vitamin D deficiency, the importance of this condition in humoral immunity will be worth looking at more closely. This study supports the role of vitamin D in the competent immune system.
Article
Patients with end-stage renal disease present an immunodeficiency that paradoxically coexists with activation of most immunocompetent cells, and the roles of chronic uremia and maintenance dialysis are poorly understood. We determined circulating levels of IL-1 beta and IL-1Ra, TNF-alpha and its soluble receptors (TNF-sR55 and TNF-sR75), and activation markers of T cells (soluble CD25), B cells (soluble CD23), and monocytes (neopterin) in a large cohort of undialyzed patients at various stages of chronic renal failure and in dialyzed patients on maintenance hemodialysis or chronic peritoneal dialysis. The progression of uremia was associated with a gradual increase in soluble CD25, CD23, and especially neopterin levels. Although IL-1 beta could not be detected, IL-1Ra levels were significantly increased from the earliest stage of renal failure. Plasma levels of TNF-alpha, TNF-sR55, and TNF-sR75 progressed with the severity of renal failure and correlated with soluble CD25, CD23, and neopterin levels, whereas IL-1Ra levels correlated exclusively with TNF-sR55 levels. Compared with undialyzed patients, levels of IL-1 beta were higher in patients on maintenance hemodialysis, whereas those of IL-1Ra were lower and decreased further at the end of dialysis sessions. In contrast, both TNF-sR55 and TNF-sR75 levels were significantly higher than in undialyzed patients and increased further at the end of dialysis sessions in the absence of an increase of TNF-alpha. Such an imbalance between cytokines and their inhibitors may play a pivotal role in the multifaceted process of immune dysfunction.
Article
The ‘classical’ effects of vitamin D receptor activator or agonist (VDRA) therapy for the treatment of secondary hyperparathyroidism in patients with chronic kidney disease primarily involves suppressive effects on the parathyroid gland, and regulation of calcium and phosphorus absorption in the intestine and mobilisation in bone. Observational studies in haemodialysis patients report improved cardiovascular and all-cause survival among those receiving VDRA therapy compared with those not on VDRA therapy. Among VDRAs, the selective VDRA paricalcitol has been associated with greater survival than nonselective VDRAs, such as calcitriol (1,25-dihydroxyvitamin D3). The survival benefits of paricalcitol appear to be linked, at least in part, to ‘nonclassical’ actions of VDRAs, possibly through VDRA-mediated modulation of gene expression. In cardiovascular tissues, VDRAs are reported to have beneficial effects such as anti-inflammatory and antithrombotic effects, inhibition of vascular smooth muscle cell proliferation, inhibition of vascular calcification and stiffening, and regression of left ventricular hypertrophy. VDRAs are also reported to negatively regulate the renin-angiotensin system, which plays a key role in hypertension, myocardial infarction and stroke. The selective VDRAs, paricalcitol and maxacalcitol, are associated with direct protective effects on glomerular architecture and antiproteinuric effects in response to renal damage. Paricalcitol regulates several cardiovascular and renal parameters more favourably than nonselective VDRAs. Complex nonclassical effects, which are not clearly understood, possibly contribute to the improved survival seen with VDRAs, especially paricalcitol.
Article
Background: The 2009 KDIGO (Kidney Disease: Improving Global Outcomes) chronic kidney disease-mineral and bone disorder clinical practice guideline suggests correcting 25-hydroxyvitamin D3 (25[OH]D) levels<30ng/mL in patients treated with maintenance hemodialysis, but does not provide a specific treatment protocol. Study design: 2-center, double-blind, randomized, 13-week, controlled trial followed by a 26-week open-label study. Setting & participants: 55 adult maintenance hemodialysis patients with 25(OH)D levels<30ng/mL were recruited from June 2008 through October 2009. Intervention: Cholecalciferol, 25,000IU, per week orally versus placebo for 13 weeks, then 26 weeks of individualized cholecalciferol prescription based on NKF-KDOQI (National Kidney Foundation-Kidney Disease Outcomes Quality Initiative) guidelines. Outcomes: Primary end point was the percentage of patients with 25(OH)D levels≥30ng/mL at 13 weeks. Secondary outcomes included the percentage of patients with normal calcium, phosphorus, and intact parathyroid hormone (iPTH) blood levels. Safety measures included incidence of hypercalcemia and hypervitaminosis D. Measurements: Blood calcium and phosphate were measured weekly; iPTH, 25(OH)D, 1,25-dihydroxyvitamin D3 (1,25[OH]2D), and bone turnover markers, trimonthly; fetuin A and fibroblast growth factor 23 (FGF-23) serum levels and aortic calcification scores were determined at weeks 0 and 39. Results: The primary end point significantly increased in the treatment group compared with the placebo group (61.5% vs 7.4%; P<0.001), as well as 1,25(OH)2D levels (22.5 [IQR, 15-26] vs 11 [IQR, 10-15]pg/mL; P<0.001) and the proportion of patients achieving the target calcium level (76.9% vs 48.2%; P=0.03). Incidence of hypercalcemia and phosphate and iPTH levels were similar between groups. The second 26-week study phase did not significantly modify the prevalence of 25(OH)D level≥30ng/mL in patients issued from the placebo group. Limitations: Small size of the study population. Conclusions: Oral weekly administration of 25,000IU of cholecalciferol for 13 weeks is an effective, safe, inexpensive, and manageable way to increase 25(OH)D and 1,25(OH)2D levels in hemodialysis patients. Further evaluation of clinical end points is suggested.
Article
Gene expression of interleukin-1β during hemodialysis. It is still controversial whether the hemodialysis (HD) procedure is an inflammatory process in vivo. Therefore, we studied the gene expression of interleukin-1β (IL-1β) as a marker of inflammation in peripheral blood mononuclear cells (PBMC) of patients during HD by Northern blotting and polymerase chain reaction. Compared to PBMC separated pre-HD (1.0 densitometric units), the amount of IL-1β mRNA was increased in PBMC leaving the dialyzer (12.2 ± 2 densitometric units, P < 0.01), but was not increased in PBMC re-entering the dialyzer from the systemic circulation (0.6 ± 0.1 densitometric units) in all 12 patients studied. The maximal amount of IL-1β mRNA in PBMC was seen at five minutes after start of HD. There was a significant correlation between the increase in IL-1β mRNA and the increase in activated complement C5a (r = 0.71, P < 0.01). HD using less complement-activating membranes (hemophan, polysulfone, polyamide or polyacrylonitrile) resulted in no detectable IL-1β mRNA. Furthermore, a monoclonal antibody against human C5a reduced the increase in IL-1β mRNA by 83% (P < 0.05), indicating that C5a plays a major role for induction of IL-1β mRNA during HD. This study demonstrates that during HD with regenerated cellulose, gene expression for IL-1β takes place in PBMC.