You are on page 1of 10

Biochimica et Biophysica Acta 1863 (2017) 907–916

Contents lists available at ScienceDirect

Biochimica et Biophysica Acta

journal homepage: www.elsevier.com/locate/bbadis

Review

Vitamin D in liver disease: Current evidence and potential directions


Harendran Elangovan a, Sarinder Chahal a, Jenny E. Gunton a,b,⁎
a
The Garvan Institute of Medical Research, The University of New South Wales (UNSW), Sydney, NSW, Australia
b
The Westmead Institute of Medical Research, The University of Sydney, NSW, Australia

a r t i c l e i n f o a b s t r a c t '

Article history: Consistent with its multifaceted nature, growing evidence links vitamin D with hepatic disease. In this review, we
Received 5 September 2016 summarise the roles of vitamin D in different liver pathologies and explore the clinical utility of vitamin D-based
Received in revised form 6 December 2016 treatments in hepatology. We find that the small number of clinical trials coupled with the profound heteroge-
Accepted 2 January 2017
neity of study protocols limits the strength of evidence needed to ascribe definite clinical value to the hormone
Available online 4 January 2017
in liver disease. Nevertheless, the experimental data is promising and further bench and bedside studies will like-
ly define a clearer role in hepatic therapeutics.
© 2017 Elsevier B.V. All rights reserved.

1. Introduction dehydrocholesterol undergoes a stereoisomeric change in response to


ultraviolet B light exposure to form pre-vitamin D3. This is influenced
While the liver is renowned for its regenerative prowess, in the face by skin pigmentation, cutaneous 7-dehydrocholesterol stores and UV
of overwhelming acute or chronic injury, these adaptive mechanisms exposure [1]. Pre-vitamin D3 is then converted to vitamin D3 via non-
may be overcome allowing progression to acute liver failure, cirrhosis, enzymatic isomerisation, a process that takes approximately 2 to
hepatic failure and/ or hepatocellular cancer (HCC). Knowledge of the 3 days and produces the bulk of vitamin D in most people.
molecular landscape of hepatic disease has burgeoned over the years. Vitamin D3 in circulation is predominantly bound to the vitamin
Consistent with its diverse properties that extend beyond the regu- D-binding protein (DBP). In the liver it undergoes a hydroxylation
lation of calcium and bone homeostasis, the vitamin D axis has received reaction to form 25(OH)vitamin D (25D). Many forms of 25-
much attention in hepatic pathophysiology. A high prevalence of vita- hydroylase have been described including CYP27A1, CYP2DII,
min D deficiency is seen in patients with liver diseases and pre-clinical CYP2D25, CYP3A4, CYP2S25, CYP2J3 and CYP2R1. CYP2R1 is sug-
evidence suggests therapeutic benefits with vitamin D-based treat- gested to play the biggest role [2] as inactivation is associated with
ments. This review summarises the roles of vitamin D in liver disease, poor hydroxylation of vitamin D and a rickets phenotype [3]. 25D un-
highlighting important mechanistic relationships and evaluates the cur- dergoes a second hydroxylation at the C1a position by 1a-hydroxy-
rent evidence for vitamin D therapeutics in the field of hepatology. lase (encoded by CYP27B1) to produce 1,25(OH)2 vitamin D
(1,25D), the biologically active hormone. Once synthesised, 1,25D
bound to DBP enters the circulation and enters target tissues to me-
2. Vitamin D metabolism diate physiological effects. While CYP27B1 is predominantly located
within the kidneys, it is increasingly recognised that many other cells
Vitamin D is a secosteroid hormone known for its critical role in and tissues express the enzymatic machinery necessary to synthe-
calcium and skeletal homeostasis. It is obtained either from dietary sise 1,25D [4]. Thus locally synthesised hormone may cause tissue-
sources or synthesised endogenously (see Fig. 1). In the skin, 7- specific autocrine and paracrine effects that are the focus of many
current research efforts.
Abbreviations: 1,25D, 1,25(OH)2 vitamin D; 25D, 25(OH) vitamin D; ACR, acute CYP24A1 inactivates 1,25D. Evidence of its importance in the process
cellular rejection; CKD, chronic kidney disease; CYP, Cytochrome P450; HBV, hepatitis B
was illustrated by the high vitamin D levels in mice lacking the enzyme,
virus; HCV, hepatitis C virus; HCC, hepatocellular cancer; HSC, hepatic stellate cell; IL,
Interleukin; I-R, ischemic-reperfusion; KLF-4, Krüppel-like factor 4; LCA, lithocholic acid; with the development of osteopetrosis [5].
NAFLD, non-alcoholic fatty liver disease; NF-kB, nuclear factor kappa beta; PBC, primary
biliary cirrhosis; RXR, retinoid X receptor; SVR, sustained virologic response; TGFβ, 3. What is hypovitaminosis D?
tumour growth factor beta; TLR, Toll-like receptor; TNFα, tumour necrosis factor alpha;
VDR, vitamin D receptor.
⁎ Corresponding author at: The Westmead Institute of Medical Research, The University
Vitamin D status is defined not by levels of the biological active
of Sydney, NSW, Australia. 1,25D but by 25D. 25D has a long half-life (approximately
E-mail address: jenny.gunton@sydney.edu.au (J.E. Gunton). 3 weeks), is stable in serum, and levels correlate to a number of

http://dx.doi.org/10.1016/j.bbadis.2017.01.001
0925-4439/© 2017 Elsevier B.V. All rights reserved.
908 H. Elangovan et al. / Biochimica et Biophysica Acta 1863 (2017) 907–916

biological endpoints [6]. In contrast, 1,25D has a short half-life lead- 4. The vitamin D receptor (VDR)
ing to more fluctuations in its level, and 1,25D levels do not correlate
well with risk of disease. VDR is a member of the nuclear hormone receptor superfamily and
Classically, vitamin D deficiency has been defined by levels at is recognised as the principle mediator of the actions of the biologically
which risk of osteomalacia and rickets increase. According to the active form of vitamin D (1,25D) or its synthetic derivatives. Bile acids
World Health Organisation, vitamin D deficiency is defined by like lithocholic acid (LCA) also endogenously activate VDR [10]. In the
25D b 10 ng/mL (25 nmol/L) whilst a level below 20 ng/mL absence of ligand, VDR was believed to be largely inactive. However,
(50 nmol/L) is deemed insufficient. However, since some people de- unliganded receptor clearly has biological roles, as shown by hair loss
velop osteomalacia at levels between 10 and 20 ng/mL, probably in- without VDR [11] and changes in brown fat physiology [12]. These find-
fluenced by calcium and phosphate intake, most experts consider a ings highlight the importance of considering this novel role of un-
level b 20 ng/mL (50 nmol/L) to be deficient. liganded VDR when evaluating receptor function.
Despite the levels above, a value of 30 ng/mL (75 nmol/L) is rec- Similar to other nuclear hormone receptors, ligand occupancy facil-
ommended as optimal because this produces the nadir in parathy- itates a conformational change which allows the receptor to interact
roid hormone levels (PTH). Finally, some recommend another with its heterodimeric partner, the retinoid X receptor (RXR) (Fig. 2).
approach defining optimal 25D as that level at which there is no fur- This ligand-heterodimeric complex subsequently migrates into the cell
ther increment in 1,25D. The conclusions from both of these ap- nucleus where it binds to target response elements within the chroma-
proaches are 30 ng/mL [7,8]. Thus, levels between 20 and 30 ng/mL tin to modulate gene transcription. Transcriptional control is further
are commonly considered insufficient. This is population data, and fine-tuned by the additional recruitment of co-regulatory molecules
optimal levels are likely to vary between individuals. The growing which manifest distinct expression profiles within different tissue
appreciation of genetic polymorphisms and/or other factors in types and thus confer pleiotropy to the VDR circuit. Ligand binding to
influencing the host response to vitamin D signaling adds another di- VDR located within plasma membrane caveolae can precipitate rapid
mension of complexity. Ideal levels may also differ across ethnic biological effects by acting via non-genomic pathways [13].
groups, and vary by the tissue being considered.
Regardless, approximately 1 billion people worldwide are be- 4.1. Vitamin D receptor in liver
lieved to have deficient 25D levels, with particularly stark statistics
noted amongst females of Middle Eastern origin [9]. Cultural factors VDR levels are low in normal liver. However, the receptor is hetero-
particularly related to dress wear and skin coverage are important. geneously distributed with low expression within unstimulated hepa-
Equatorial countries that classically experience high levels of ultravi- tocytes and robust expression within biliary-epithelial, Kupffer and
olet radiation year-round are now showing increasing rates of defi- other non-parenchymal cell types [14]. This distribution suggests that
ciency [9]. In these cases, factors such as obesity and sedentary the liver may initially respond to vitamin D through its non-parenchy-
lifestyle, darker skin pigmentation, use of sunscreen and UV avoid- mal components. Ding and colleagues [15] recently elucidated a VDR/
ance may explain this trend. It is important to note here that sun- SMAD genomic circuit in stellate cells which strongly antagonises
avoidance strategies have led to significant decreases in skin cancer TGF-β signaling, the most potent pro-fibrogenic pathway in the liver.
rates, so there is an important ‘trade-off’ between UV exposure and VDR null mice have increased susceptibility to cholestatic injury [16]
skin cancer versus adequate vitamin D. mediated through disruptions in biliary epithelial junctions, distortions

Fig. 1. Vitamin D synthesis and metabolism. Vitamin D synthesis largely occurs endogenously with sunlight exposure. It is then hydroxylated, first in the liver and then in the kidney, to
produce 1,25D. This metabolite is biologically active and mediates a range of responses across different tissues in the body. 1,25D is inactivated by the action of CYP24A1.
H. Elangovan et al. / Biochimica et Biophysica Acta 1863 (2017) 907–916 909

in bile acid homeostasis and a diminution in ductal reaction which func- virologic response (SVR) [31,32]. Kitson et al. [31] evaluated 11 studies
tions as an adaptive drainage mechanism. The latter two effects are he- and concluded there were no significant associations between vitamin
patocellular in origin and the authors postulated possible indirect D status and SVR in response to Peg-Interferon/Ribavirin therapy. In
influences mediated through vitamin D actions upon target non-paren- contrast, the meta-analysis by Garcia-Alvarez and colleagues [32] con-
chymal or extra-hepatic cells [16]. However, a direct role has been cluded that low vitamin D (b20 ng/mL) was linked to a lower odds of
proposed for the non-classical VDR pathway in cultured human hepato- achieving SVR. However, this report had several limitations including
cytes which acts to inhibit bile acid synthesis and cholestasis [13]. We key study omissions and inclusion of 3 papers which utilised the same
note that the process of cell isolation and passaging in culture can itself cohort of Italian subjects. Their findings were no longer significant
induce VDR [17]. Hence, in vitro approaches cannot definitively clarify once two out of the three Italian studies were excluded [33]. Since
levels of hepatocellular VDR in vivo. then, two recent studies by Belle et al. [34] and Loftfield et al. [35] did
Despite the evidence implicating the vitamin D axis in hepatic biol- not identify any beneficial associations between baseline vitamin D
ogy, Wang, Deluca and colleagues were unable to visualise VDR in level and SVR. Loftfield and colleagues [35] found that high 25D
HSCs and other non-parenchymal cells [18]. However, in another tissue (≥30 ng/mL) was associated with lower odds of early virologic response
(muscle) reported not to have VDR expression, Girgis et al. [19] demon- to treatment [36]. Overall, baseline levels of 25D do not predict out-
strated with the same antibody that VDR is readily detectable if a high- comes with older anti-viral treatments. Consistent with this, Terrier et
salt lysis buffer is used. VDR is a ‘sticky’ protein, and use of high-salt lysis al. [37] failed to identify any beneficial impact associated with
buffer or more stringent antigen retrieval may modulate the amount of correcting vitamin D levels prior to the initiation of Peg-interferon/riba-
‘free’ VDR which can bind antibody. Regardless, it is clear that VDR ex- virin in previous non-responders with HCV genotype 1 or 4 infection.
pression is low in normal liver.
However, for many transcription factors, low levels are sufficient to
5.1.1. Genetic polymorphisms, vitamin D and HCV
mediate function. The current difficulties surrounding VDR detection
Studies have focussed upon vitamin D related gene polymorphisms
highlight the importance of alternative methodologies such as targeted
and their potential roles in influencing treatment outcome with both
genetic knockdown as a complementary means to establish proof of in
Baur et al. [38] and Garcia-Martin et al. [39] describing that VDR bAt
vivo VDR functionality. For example, deletion of VDR from
[CCA] haplotype impairs the antiviral response to Peg-interferon/ribavi-
cardiomyocytes has provided much useful insight into the importance
rin treatment in Caucasian patients with chronic hepatitis C. In contrast,
of cardio-myocellular vitamin D signaling pathways upon the mainte-
a newer study by Hung et al. [40] found no significant associations be-
nance of heart health in vivo [20].
tween various VDR polymorphisms, including the aforementioned bAt
Another intriguing concept is the notion of VDR upregulation sec-
[CCA] haplotype and treatment response in an Asian cohort of chronic
ondary to a pathologic stimulus (Fig. 3). This would be consistent with
hepatitis C patients. The authors attributed the discordant results to
the observations of VDR expression/activity in transformed hepatocytes
possible ethnic differences in vitamin D physiology/handling [40]. The
[21,22] and the inverse correlations between VDR levels and severity of
authors also highlighted the higher prevalence of a favourable Interleu-
non-alcoholic fatty liver disease described by Barchetta et al. [23]. Ab-
kin (IL) 28B polymorphism in Asian populations which promotes SVR
sence of appropriate positive and negative controls in these studies
[40]. These findings were mirrored in recent study by Arai et al. [41]
complicates interpretation of signal validity. Nevertheless, lessons
who found that polymorphisms in various genes involved in vitamin
from the study of skeletal muscle suggest that VDR expression is not a
D handling could predict neither vitamin D levels nor SVR in a Japanese
static process with Girgis and colleagues [19] reporting VDR downregu-
cohort of HCV genotype 1b patients. There are therefore no consistent
lation as myoblasts mature into myotubes, and that young mice have
effects across different polymorphisms for effect on HCV.
higher receptor content within their skeletal muscle than older mice.

5. Vitamin D and chronic hepatitis C 5.1.2. Vitamin D supplementation and HCV


Although baseline levels of 25D do not appear to predict outcome in
Approximately 160 million people worldwide are chronically infect- HCV infection, there are some promising results for supplementation.
ed with the hepatitis C virus (HCV), and it carries an increased risk of Two small randomised studies from Israel showed that subjects infected
liver cirrhosis and HCC [24]. The newer anti-virals offer a major advance, with HCV genotypes 1–3 who received vitamin D supplementation
but the role of vitamin D with these agents is largely unknown. Clearly, targeting a 25D level N 80 nmol/L in addition to standard Peg-interfer-
more data on the new regimens is required, but given their high effica- on/ribavirin manifested improved outcomes compared to controls
cy, further increments with vitamin D would require very large num- who received only the IFN-based therapy [42,43]. Recipients of vitamin
bers to detect, and these studies are not likely to be done. D supplementation had improved indices of insulin resistance, sug-
Vitamin D deficiency appears common in HCV patients [25,26]. In gested to be a factor predicting response to Peg-interferon/ribavirin
the first study, 28/43 (65%) of patients with HCV and cirrhosis had [44]. Similar findings were elicited in a study by Yokoyama and col-
25D b 20 nmol/L and 49/57 (86%) of people with HCV without cirrhosis leagues [45], who examined IL-28 polymorphisms in predicting re-
were b20 nmol/L [25]. Mean vitamin D was deficient at 17 ng/mL in a sponse to vitamin D supplementation. However, not all studies are
study of 468 HCV patients [26]. positive with Ladero et al. [46] reporting no significant effect of vitamin
Current evidence suggests that vitamin D might play a role in HCV D supplementation alone upon HCV viral load or biochemical markers
infection persistence and clearance. Observations from in vitro studies of necro-inflammation. It is possible that the 5–7 week duration of
suggest that the hormone suppresses HCV replication through the in- that study may have been insufficient to detect a difference. In a group
duction of oxidative stress pathways [27], promotion of interferon- of 100 HCV genotype 4 patients, Esmat et al. [47] reported no benefit
based signaling, and increases in chemotactic factors and host autopha- of vitamin D supplementation upon SVR.
gic machinery [28,29]. Vitamin D also enhances zinc uptake [28], and To summarise, there is in vitro data suggesting decreased viral repli-
zinc is a negative regulator of HCV replication [30]. cation with vitamin D treatment. There are no data suggesting influence
of baseline vitamin D on outcome, however, treating at the time of anti-
5.1. Baseline vitamin D and HCV infection viral therapy may beneficially influence outcomes. We suggest that peo-
ple with 25D levels b 30 ng/mL should be treated to N30 ng/mL as this
However, the clinical literature is more controversial. Two recent may positively influence HCV, and is likely to be beneficial for bone
meta-analyses reached conflicting conclusions about the ability of low and calcium homeostasis. The evidence is insufficient to recommend
baseline vitamin D to predict the degree of injury/fibrosis and sustained treating people whose level is normal (N30 ng/mL).
910 H. Elangovan et al. / Biochimica et Biophysica Acta 1863 (2017) 907–916

Fig. 2. The vitamin D/VDR signaling axis. Classically, liganded VDR undergoes a conformational change which facilitates binding with RXR. This complex migrates to the nucleus and binds
to vitamin D response elements within the DNA to regulate gene transcription. This action is modulated by a range of transcriptional co-regulators. Emerging evidence implicates a subset
of membrane-bound VDR in the triggering of rapid biological actions via non-genomic pathways. Unliganded VDR is physiologically active, and modulates brown fat and hair follicle
physiology.

5.2. Vitamin D and chronic hepatitis B described by Liu et al. [57]. Further studies evaluating these genetic
polymorphisms are warranted. As exemplified by the HCV experience,
Similar to the results for HCV, Farnik et al. [48] highlighted that low the effect of VDR gene polymorphisms upon the course of HBV infection
25D levels are linked with higher viral replication rates in patients with may differ between different ethnicities. Nevertheless, the early data
chronic HBV infection, results reflected in a recent Vietnamese study of suggests the possibility of vitamin D focussed pharmacogenomics to
400 HCV patients by Hoan and colleagues [49]. Both studies reported predict treatment response and/or tailor therapeutic regimens.
that vitamin D deficiency was common in people with HBV infection.
In contrast, neither Zhao et al. [50] did not identify a relationship be-
5.2.1. HBV vaccination
tween vitamin D and HBV DNA. Chan et al. [51] identified a significant
Renal failure impairs the immune response and some authors have
relationship between baseline vitamin D levels and HBV DNA but this
examined the relationship between vitamin D and HBV vaccination.
was no longer significant after correction for age, gender, HBeAg and
Zitt et al. [58] found that low vitamin D levels (i.e. b10 ng/mL) in chronic
HBV genotype. As suggested by Chan et al. [52], the discordant findings
kidney disease (CKD) patients predicted a poor response to vaccination.
may be explained by the idea that the division of the chronic HBV phe-
In contrast, Jhorawat and colleagues [59] reported that vitamin D levels
notype into distinct clinical stages reflects changing between the virus
did not differ between vaccine responding and non-responding dialysis
and host immunity which may respond differently to vitamin D signal-
patients. Vitamin D levels tended to be higher in responders, and the au-
ing, or that the association runs in the other direction: worsened liver
thors suggested that the small sample size and significant loss to follow
disease causes lower 25D. Elucidating the temporal course of 25D levels
up may account for their findings.
over the natural history of chronic HBV infection may provide further
We were not able to identify any published studies about HBV vacci-
information.
nation efficacy and vitamin D in people without renal failure. Further
Studies have examined the role of host vitamin D genetics in
examination of vaccine response is warranted, but it is prudent to en-
influencing the outcomes of HBV infection. Boglione et al. [53] elucidat-
sure normal vitamin D status in renal patients to avoid the extra stimu-
ed that the TT genotype of the CYP27B1 + 2838 single nucleotide poly-
lus to hyperparathyroidism that deficiency provokes.
morphism (SNP) was predictive of favourable virologic outcomes in
patients treated with PEG-IFN. Associations between distinct clinical
phenotypes and VDR polymorphisms are described in Taiwanese hepa- 6. Vitamin D and non-alcoholic fatty liver disease
titis B virus (HBV) carriers [54], as have links between Taq1 and Fok1
polymorphisms, and infection-related outcomes in Chinese patients NAFLD includes a spectrum of disorders of increasing severity with
[55]. Bellamy et al. [56] found a single T to C base change polymorphism liver lipid deposition, inflammation, fibrosis and cirrhosis. It is predicted
in codon 352 of VDR to be correlated with diminished rates of persistent to become the most common cause of cirrhosis in developed countries
HBV and tuberculosis. The improved response in these patients may be as HBV and HCV infection rates are brought under control with vaccina-
analogous to the vitamin D mediated cathelicidin anti-TB defence tion (HBV) and antivirals.
H. Elangovan et al. / Biochimica et Biophysica Acta 1863 (2017) 907–916 911

Fig. 3. The VDR may be upregulated in response to hepatic injury.

Vitamin D deficiency is more common in people with NAFLD [23,60]. 6.2. Preclinical human data
Clinical studies in adult and pediatric populations suggest an inverse as-
sociation between 25D and the histologic severity of NAFLD. A recent The hepatic transcriptome of NAFLD patients shows enhanced acti-
meta-analysis of 29 case-control and cross-sectional studies [61] vation of mitogen-activated protein kinase and NF-κB related pro-in-
found that patients with NAFLD were 26% more likely to be vitamin D flammatory signaling pathways [75]. Challenging cultured adipocytes
deficient than controls. These findings were corroborated in another with vitamin D promotes adiponectin secretion [73]. Adiponectin has
meta-analysis by Eliades et al. [62] and in a recent large Korean study potent anti-inflammatory and insulin sensitising properties, including
by Chung and colleagues [63]. However, the various primary studies sig- in the liver, and its levels are reduced in NAFLD patients [76–78]. Of
nificant heterogeneity in the definition and determination of vitamin D note, Miele et al. [79] reported that a leaky gut was correlated with in-
levels, and the criteria utilised to diagnose NAFLD. It is important to note creased steatosis in NAFLD patients, supporting the involvement of
that not all data s supportive; Bril et al. [64] report that vitamin D levels entero-hepatic interactions in the pathogenesis of metabolic injury.
were not associated with NAFLD severity in 239 biopsy samples. These An elegant study utilising radiolabelled isotopes to track lipid migra-
findings are in concordance with other data that utilised similar biop- tion determined that the majority of intrahepatic triglyceride originates
sy-based protocols [65–67]. from dietary fat [80]. Given the known importance of bile acids in lipid
While it is tempting to suggest that liver disease may impair process- metabolism, coupled with the emerging roles of the vitamin D axis as a
ing of vitamin D to 25D, hepatic expression of both CYP2R1 and negative regulator of biliary homeostasis [13,16], it is possible that defi-
CYP27A1 are preserved in patients with steatohepatitis [23]. These find- ciency might promote lipid uptake. Ning and colleagues [81] also re-
ings were corroborated by Patel et al. [67] who further identified that cently showed that vitamin D administration also protects against
the expression of key CYP genes involved in vitamin D metabolism did steatosis by stimulating increasing peroxisome proliferator activated re-
not vary with increasing severity of NAFLD. These observations suggest ceptor alpha (PPARα) mediated lipolysis and lipid clearance.
that loss of vitamin D hydroxylation capacity is not the cause of
hypovitaminosis D in NAFLD patients. It is conceptually obvious that
6.3. Clinical studies in non-alcoholic fatty liver disease
low vitamin D levels may be a consequence of shared risk factors for
NAFLD and low vitamin D such as a sedentary lifestyle.
Despite the encouraging pre-clinical studies, interventional studies
in NAFLD are largely negative. Barchetta et al. [82] found no difference
in hepatic fat fraction and fatty liver index in 55 patients with type 2 di-
6.1. Pre-clinical data: animal models
abetes and NAFLD given 2000 IU per day of vitamin D for 6 months.
However, we note that the baseline numbers for fatty liver index in
Studies report links between low vitamin D, insulin resistance and the 2 groups of patients are not the same in Tables 1 and 3, making
hepatic inflammation. Vitamin D deficiency exacerbates liver inflamma- data interpretation challenging. Given the large standard deviations,
tion in a rodent model of NAFLD, findings ascribed to increased insulin the study was not adequately powered to detect a modest change in
resistance and the upregulation of pro-inflammatory genes including these measures.
resistin, Tnfα, Il-4, Il-6 and the Toll-like receptors (Tlr) [68]. Photothera- High-dose oral vitamin D3 supplementation (25,000 IU/week) over
py, which increases vitamin D, attenuated the hepatic inflammatory and 24 weeks had no beneficial impact upon liver histology, hepatic bio-
fibrotic milieu whilst improving markers of insulin resistance in animal chemical indices, insulin resistance or adipokine profile in 12 non-dia-
models [69]. The details underpinning these hepato-protective effects betic patients with biopsy-proven steatohepatitis but again this study
are poorly understood. Observations that insulin release by pancreatic was not powered to detect moderate differences [83].
beta cells appears to be a vitamin D-responsive phenomenon [70,71], Sharifi et al. [84] reported that high dose supplementation
VDR null mice have impaired insulin secreting capacity [70], and vita- (50,000 IU fortnightly for 4 months in 27 patients) decreased
min D can ameliorate insulin resistance through the promotion of glu- inflammatory markers such as serum C reactive protein and
cose uptake in myocytes [72] and adipocytes [73] suggest that there malondialdehyde in NAFLD compared to 26 people who received place-
may be many contributing mechanisms. bo, although they observed no beneficial effects upon ultrasonographic
As illustrated in the whole body VDRKO mouse, vitamin D promotes and or other markers of hepatic injury. Again, study is probably not ad-
CD8aa T cell maturation, cells implicated in the regulation of enteric im- equately powered to detect changes in ultrasound measures.
munity [74]. Intriguingly, dysregulated gut immunity may contribute In summary, although in vivo and in vitro data link hypovitaminosis
towards hepatic pathology since disruption of the intestinal barrier D with NAFLD, a causative link in either direction remains unproven.
may facilitate translocation of antigenic substrates such as bacterial Appropriately powered randomised trials with standardisation of
products into the hepatic microenvironment, triggering immune study methodology and outcome measures are needed in order to
responses. determine the effect of vitamin D supplementation in NAFLD.
912 H. Elangovan et al. / Biochimica et Biophysica Acta 1863 (2017) 907–916

7. Vitamin D and autoimmune liver disease reverse TNFα mediated downregulation of CD28 in liver-infiltrating T
cells, a process believed to exacerbate the inflammatory phenotype
Consistent with the links between vitamin D and autoimmunity, vi- and is linked to the pathogenesis of PSC.
tamin D signaling may be important in primary biliary cirrhosis (PBC)
[85–89]. Genome wide association analyses in PBC patients demonstrat-
ed that VDR Bsm1 and Taq1 polymorphisms were associated with sus- 8. Vitamin D and hepatic transplantation
ceptibility to advanced fibrosis/cirrhosis [90]. In recent years, studies
by both Guo et al. [91] and Agmon-Levin et al. [92] identified that low As an extension of the links between vitamin D and hepatic im-
vitamin D levels were significantly correlated with major indices of he- munity, a few studies have explored the potential for vitamin D to
patic dysfunction in PBC patients. Importantly, the Guo paper highlight- modulate transplant outcomes. Redaelli and colleagues [94] found
ed that baseline vitamin D levels predicted the subsequent response to in a rodent model of liver transplantation that 1,25D in combination
ursodeoxycholic acid therapy. with low-dose cyclosporine decreased the severity of the acute cellu-
As highlighted previously, both Firrincieli et al. [16] and Han et al. lar rejection (ACR) and prolonged survival of hepatic allografts. Sim-
[13] provided mechanistic insight into the ways vitamin D signaling ilarly, Xing et al. [95] recently reported that calcitriol ameliorates
might contribute to the cholestatic phenotype. Moreover, many compo- ACR in liver transplants, findings the authors ascribed to circulating
nents of the immune system are vitamin D sensitive. Vitamin D signal- T-regulatory cells. Association between specific VDR polymorphisms
ing directs dendritic cell migration, alters the cytokine secretome and and ACR was shown by Falleti and colleagues [96]. These findings are
promotes the development of self-tolerance [52]. The hormone may consistent with the observation that certain polymorphic loci are
temper the adaptive immune response with Boglione and colleagues sited near the locale of CpG islands [97] which through site-specific
[53] highlighting potential functions in the regulation of B cell prolifer- methylation, can affect transcription factor binding and gene expres-
ation, differentiation and autoantibody production. Finally, vitamin D sion. Experimental models suggest that VDR allelic heterogeneity
also functions as an inhibitor of Th1 and Th17 responses whilst increas- can exert influence upon receptor expression levels, which would
ing production of IL-10, T-regulatory cells and Natural Killer T cells, ef- likely impact upon any immunomodulatory effects of the vitamin
fects implicated in the pathogenesis of autoimmunity [54]. D/VDR signaling axis [98].
Comparatively less work has been performed in the domains of pri- Further research is merited in order to more clearly define a role for
mary sclerosing cholangitis (PSC) and autoimmune hepatitis. However, vitamin supplementation in the transplant population. However, with
both Vogel et al. [87] and Fan et al. [85] described associations between the markedly increased risk of fractures post-transplantation, it is im-
VDR Fok polymorphisms and autoimmune hepatitis. More recently, in portant to treat all people who are vitamin D deficient in this population
vitro work by Liaskou and colleagues [93] suggests that vitamin D can to normal levels.

Fig. 4. Summary of the actions of vitamin D across the spectrum of liver disease. The vitamin D/VDR axis plays a critical role in modulating the onset and severity of a range of hepatic
insults. In acute liver injury, vitamin D is suggested to block the activation of Kupffer macrophages via the suppression of TLR-4 signaling. The actions of vitamin D in NAFLD are
typified by the down-regulation of inflammatory cytokines and in the amelioration of insulin resistance. Vitamin D's role as a negative regulator of biliary homeostasis may help
protect against steatosis and cholestatic injury. Vitamin D modulates fibrosis by displacing Smad binding. This signaling pathway is suggested to play a role in hepatic carcinogenesis.
Vitamin D suppresses tumour proliferation by inhibiting C-myc. In viral hepatitis, vitamin D promotes interferon-dependent pro-inflammatory signaling which causes an antiviral
effect. Vitamin D has manifold effects upon immune cells and self-tolerance.
H. Elangovan et al. / Biochimica et Biophysica Acta 1863 (2017) 907–916 913

9. Vitamin D and hepatic fibrosis property of vitamin D is specific to mTOR inhibitors or can be more
broadly applied. Moreover, Bromodomain and Extra-Terminal motif
In recent years, there has been burgeoning interest in the utility of (BET) inhibitors have recently been shown to suppress Myc functional-
vitamin D as a potential anti-fibrotic agent in chronic liver disease. Cen- ity in experimental cancer models [110].
tral to this hypothesis are findings by Ding and colleagues [15], who re- However, novel findings by Chen et al. [111] suggests that vitamin D
cently elucidated that that the vitamin D-VDR complex can function as a deficiency might enhance hepatic carcinogenesis in rodent models with
ligand-dependent transcriptional repressor within HSCs, antagonising disrupted TGFβ/Smad-3 signaling through the upregulation of TLR-7
TGFβ mediated recruitment of SMAD3 to the regulatory loci of numer- and β-catenin/Wnt signaling networks. Intriguingly, the authors also
ous pro-fibrotic genes including collagens, integrins and tissue inhibi- showed that vitamin D treatment restored TGFβ pathway proteins
tors of metalloproteinases. This TGFβ/SMAD pathway within HSCs is and suppressed β-catenin expression in both cirrhotic and HCC patients.
one of the most potent pro-fibrogenic pathways within the liver and These results are surprising as they would seem inconsistent with the
the administration of vitamin D a synthetic analogue calcipotriol can previously described role of vitamin D as an antagonist of fibrogenesis
prevent/abrogate the fibrotic response in different rodent models of via TGFβ/SMAD network attenuation [15]. Nevertheless, just as TGFβ
chronic hepatic injury [15,99,100]. signaling is suggested to manifest pleiotropic functions in hepatic dis-
The ability of vitamin D supplementation to remediate established ease, it is possible that the vitamin D/VDR axis might demonstrate a
fibrosis is less promising, with no beneficial impact in a thioacetamide broad phenotype dictated by pathological context and target cells.
model of cirrhosis [101]. These findings were recapitulated in a recent This could account for the findings by Ethier et al. [112] that vitamin D
randomised control trial (n = 36) of cirrhotic patients [102], the first deficiency independent of calcium status retards the regeneration in ro-
of its kind which tested 8 weeks of 2800 IU vitamin D supplementation dents subject to two-thirds partial hepatectomy, ascribing hepatocellu-
without detected benefit. The disappointing findings may be related to lar mitogenic properties to the hormone.
small sample size, the short study duration, or lack of effect. Consistent with data implicating the fibrotic microenvironment as a
Studies suggest that pathologic biliary compounds like lithocholic tumourigenic niche, there is growing interest in the idea that the anti-fi-
acid, which accumulate in the setting of cholestasis, can alter vitamin brotic potential of the vitamin D axis may be directed to effect chemo-
D signaling [103]. As bile acids can activate VDR, this highlights the fas- therapeutic benefits as well. Fascinating work in pancreatic ductal
cinating notion of ligand-specific pleiotropy within VDR networks adenocarcinoma found that calcipotriol treatment induced quiescence
where different ligands (i.e. vitamin D and bile acids) may converge in pancreatic stellate cells, with inhibition of tumour supportive stromal
upon common pathways to promote potentially opposing effects. signaling and increased intra-tumoural gemcitabine bioavailability
Novel work also highlights the important role of the signaling adapter [113]. These alterations resulted in diminished tumour volume and
and autophagy substrate, p62 in facilitating the stability of the VDR/ marked improvements in indices of survival over standard gemcitabine
RXR heterodimer, promoting target gene transcription [104]. Converse- monotherapy in rodent models [113]. In light of this data, the emerging
ly, loss of p62 in HSCs impairs the repression of fibrogenesis by vitamin roles of p62 in vitamin D dependent HSC regulation [104] thus high-
D ligands [104]. The activation state of p62 in target cells might thus lights its utility as a combined biomarker to predict response to VDR ac-
represent a piece of the mechanistic. The data predicts that patients tivators in the setting of both fibrosis and HCC.
with low levels of p62 in HSCs will be poor responders to vitamin D
treatments, underscoring the need to consider p62 expression profiles 10.1. Clinical studies
as a key criterion for patient stratification in future intervention studies.
Several questions remain unanswered. Are supra-physiologic doses The clinical efficacy of vitamin D in HCC has only enjoyed modest in-
needed to achieve benefit in liver fibrosis or is vitamin D ineffective in vestigation to date, with mixed results. A 2002 phase I trial involving the
late disease? Vitamin D might demonstrate a greater impact upon the use of 1,25D dissolved in 5 mL of lipiodol injected directly into the he-
initiation stages rather than progression of liver fibrosis in which case, patic artery of 8 patients with refractory HCC yielded no clear survival
could intervention be effective if given earlier? Regardless, as described improvements [114]. Three of the patients in the study developed hy-
for other conditions above, cirrhosis is associated with increased risk of percalcemia, although this was not dose limiting.
fracture, and people with cirrhosis and vitamin D deficiency should be A Phase II clinical trial involving 56 patients using EB1089 demon-
treated to achieve a normal level. strated tumour shrinkage and disease stabilisation in 2 and 12 (of 33
evaluated) patients respectively [115]. However, the small sample size
10. Vitamin D and hepatocellular carcinoma (HCC) and absence of controls make it difficult to assess benefit. Adequately-
powered human trials with appropriate controls and parameters for
Vitamin D treatment is associated with anti-proliferative activity in disease outcome are needed.
various HCC lines [22]. Consequently, efforts to exploit this have result- Bioavailability considerations in the injured microenvironment is an
ed in the development of vitamin D analogues such as EB1089, CB 1093 interesting line of inquiry and it will be useful to identify if more
and MART-10, which demonstrate significantly enhanced mito-inhibi- targeted delivery of vitamin D or its analogues can produce improved
tory properties when applied to in vitro and animal xenograft models, chemotherapeutic outcomes. The utility of vitamin D as a complement
while ameliorating the hypercalcemia seen with vitamin D [105–107]. to prevailing medical and/or surgical paradigms is also poorly under-
Intriguingly, EB1089 exhibited chemo-prophylactic properties against stood and warrants further evaluation. It is worth exploring if combina-
HCC development in C3H/Sy mice, a rodent strain notable for its suscep- tion therapy with vitamin D can yield any clinical benefits in the setting
tibility to spontaneous liver carcinogenesis [108]. Dysregulated of HCC.
Krüppel-like factor 4 (KLF-4) signaling attenuates VDR expression with- The idea is theoretically appealing. While an abrogation of the fibrot-
in HCC cell lines, diminishing the anti-proliferative effects of vitamin D ic response would in and of itself be a landmark achievement, it alone
[21]. This suggests that KLF-4 expression profiles might predict treat- may be insufficient to change the oncogenic destiny. While the chemo-
ment response to vitamin D-based therapies in HCC. therapeutic property of p62 is ascribed to its ability to potentiate vita-
Pivonello and colleagues [109] identified that vitamin D administra- min D mediated HSC shutdown [104], work by Umemura et al. [116]
tion can reverse everolimus resistance in JHH-6 HCC cells. The authors suggests that p62 promotes carcinogenesis when overexpressed in he-
reported that vitamin D downregulated C-myc levels, a potent cellular patocytes. Given the recently identified interactions between p62 and
mitogen. While still preliminary, the results of this study suggest that vi- vitamin D signaling, can the hormone contribute to hepatic oncogenesis
tamin D can potentially be exploited to expand our repertoire of chemo- as well? What are the factors that decide which pathway predominates
therapies for HCC. It will be intriguing to determine if this “revitalising” at any given time? Where do the findings by Chen et al. [111] detailing
914 H. Elangovan et al. / Biochimica et Biophysica Acta 1863 (2017) 907–916

the role of the vitamin D axis in restoring TGF pathway proteins fit into [7] R.P. Heaney, Functional indices of vitamin D status and ramifications of vitamin D
deficiency, Am. J. Clin. Nutr. 80 (2004) 1706S–1709S.
this entire molecular landscape? [8] M.C. Chapuy, P. Preziosi, M. Maamer, S. Arnaud, P. Galan, S. Hercberg, P.J. Meunier,
Prevalence of vitamin D insufficiency in an adult normal population, Osteoporos.
11. Vitamin D and acute liver injury Int. 7 (1997) 439–443.
[9] C. Palacios, L. Gonzalez, Is vitamin D deficiency a major global public health prob-
lem? J. Steroid Biochem. Mol. Biol. 144PA (2014) 138–145.
The hepato-protective properties of vitamin D may extend to acute [10] M. Makishima, T.T. Lu, W. Xie, G.K. Whitfield, H. Domoto, R.M. Evans, M.R. Haussler,
liver injury. Seif and Abdelwahad [117] demonstrated that pre-treat- D.J. Mangelsdorf, Vitamin D receptor as an intestinal bile acid sensor, Science 296
(2002) 1313–1316.
ment with vitamin D ameliorated necro-inflammatory and apoptotic [11] S.M. Lee, J.J. Goellner, C.A. O'Brien, J.W. Pike, A humanized mouse model of hered-
stigmata following the induction of hepatic ischemic-reperfusion (I-R) itary 1,25-dihydroxyvitamin D-resistant rickets without alopecia, Endocrinology
injury in rats, findings that were reproduced in a similar model of I-R in- 155 (2014) 4137–4148.
[12] P.J. Malloy, B.J. Feldman, Cell-autonomous regulation of brown fat identity gene
jury performed by Kim and colleagues [118]. There are no human clini-
UCP1 by unliganded vitamin D receptor, Mol. Endocrinol. 27 (2013) 1632–1642.
cal trials reported in this space. [13] S. Han, T. Li, E. Ellis, S. Strom, J.Y.L. Chiang, A novel bile acid-activated vitamin D re-
The above studies suggest that vitamin D supplementation can ceptor signaling in human hepatocytes, Mol. Endocrinol. 24 (2010) 1151–1164.
[14] M. Gascon-Barre, C. Demers, A. Mirshahi, S. Neron, S. Zalzal, A. Nanci, The normal
prime the liver to withstand insults. The authors attributed the
liver harbors the vitamin D nuclear receptor in nonparenchymal and biliary epi-
favourable response manifest in these vitamin D supplemented rodents thelial cells, Hepatology 37 (2003) 1034–1042.
to attenuated TLR-4 signaling, which in complex with free fatty acids [15] N. Ding, Ruth T. Yu, N. Subramaniam, Mara H. Sherman, C. Wilson, R. Rao, M.
can function as a potent activator of Kupffer cells. As reported previous- Leblanc, S. Coulter, M. He, C. Scott, Sue L. Lau, Annette R. Atkins, Grant D. Barish,
Jenny E. Gunton, C. Liddle, M. Downes, Ronald M. Evans, A vitamin D receptor/
ly, aberrant fatty-acid/TLR-4 signaling is implicated in the pathogenesis SMAD genomic circuit gates hepatic fibrotic response, Cell, 153 601–613.
of NAFLD [68], elucidating common themes perpetuating acute and [16] D. Firrincieli, S. Zúñiga, C. Rey, D. Wendum, E. Lasnier, D. Rainteau, T. Braescu, T.
chronic liver disease. I-R injury is an important consideration in hepatic Falguières, M. Boissan, A. Cadoret, C. Housset, N. Chignard, Vitamin D nuclear re-
ceptor deficiency promotes cholestatic liver injury by disruption of biliary epithe-
surgery where vascular manipulations are common, highlighting the lial cell junctions in mice, Hepatology 58 (2013) 1401–1412.
utility of peri-operative vitamin D pre-conditioning as a potentially sim- [17] A.K. Bhalla, W.C. Wojno, M.B. Goldring, Human articular chondrocytes acquire
ple and cost-effective initiative to improve patient outcomes. Studies 1,25-(OH)2 vitamin D-3 receptors in culture, Biochim. Biophys. Acta 931 (1987)
26–32.
addressing this clinical question are warranted. The limited data in the [18] Y. Wang, J. Zhu, H.F. DeLuca, Where is the vitamin D receptor? Arch. Biochem.
domain of acute/fulminant hepatic pathology emphasises the need for Biophys. 523 (2012) 123–133.
more proof-of-concept studies before vitamin D therapies can be advo- [19] C.M. Girgis, N. Mokbel, K.M. Cha, P.J. Houweling, M. Abboud, D.R. Fraser, R.S. Mason,
R.J. Clifton-Bligh, J.E. Gunton, The vitamin D receptor (VDR) is expressed in skeletal
cated in the setting of acute liver injury.
muscle of male mice and modulates 25-hydroxyvitamin D (25OHD) uptake in
myofibers, Endocrinology 155 (2014) 3227–3237.
12. Conclusions [20] S. Chen, C.S. Law, C.L. Grigsby, K. Olsen, T.T. Hong, Y. Zhang, Y. Yeghiazarians, D.G.
Gardner, Cardiomyocyte-specific deletion of the vitamin D receptor gene results in
cardiac hypertrophy, Circulation 124 (2011) 1838–1847.
Our expanding understanding of the mechanisms through which vi- [21] Q. Li, Y. Gao, Z. Jia, L. Mishra, K. Guo, Z. Li, X. Le, D. Wei, S. Huang, K. Xie, Dysregu-
tamin D and VDR modulate liver homeostasis implicates vitamin D in a lated Krüppel-like factor 4 and vitamin D receptor signaling contribute to progres-
wide spectrum of hepatic disease (Fig. 4). Despite exciting in vitro and sion of hepatocellular carcinoma, Gastroenterology 143 (2012) 799–810, e792. .
[22] M.H. Pourgholami, J. Akhter, Y. Lu, D.L. Morris, In vitro and in vivo inhibition of liver
in vivo data, therapeutic prospects in liver disease remain controversial cancer cells by 1,25-dihydroxyvitamin D3, Cancer Lett. 151 (2000) 97–102.
given the relative paucity of well-powered randomised controlled trials, [23] I. Barchetta, S. Carotti, G. Labbadia, U.V. Gentilucci, A.O. Muda, F. Angelico, G.
methodological heterogeneity and challenges in defining an optimal Silecchia, F. Leonetti, A. Fraioli, A. Picardi, S. Morini, M.G. Cavallo, Liver vitamin D
receptor, CYP2R1, and CYP27A1 expression: relationship with liver histology and
serum level of vitamin D for the liver. The data illustrates the intriguing vitamin D3 levels in patients with nonalcoholic steatohepatitis or hepatitis C
notion that the ideal hormonal level may vary between different tissues virus, Hepatology 56 (2012) 2180–2187.
and diseases. The experimental data is promising for some hepatic pa- [24] D. Lavanchy, Evolving epidemiology of hepatitis C virus, Clin. Microbiol. Infect. 17
(2011) 107–115.
thologies, including treatment with ‘calcium-sparing’ vitamin D ana- [25] J. Arteh, S. Narra, S. Nair, Prevalence of vitamin D deficiency in chronic liver disease,
logues. Because of links between liver disease and increased risk of Dig. Dis. Sci. 55 (2010) 2624–2628.
bone fractures, we recommend that people with vitamin D deficiency [26] C.M. Lange, J. Bojunga, E. Ramos-Lopez, M. von Wagner, A. Hassler, J. Vermehren, E.
Herrmann, K. Badenhoop, S. Zeuzem, C. Sarrazin, Vitamin D deficiency and a
should be treated to achieve normal 25D (N 30 ng/mL). CYP27B1-1260 promoter polymorphism are associated with chronic hepatitis C
and poor response to interferon-alfa based therapy, J. Hepatol. 54 (2011) 887–893.
Transparency document [27] M. Yano, M. Ikeda, K.-i. Abe, H. Dansako, S. Ohkoshi, Y. Aoyagi, N. Kato, Comprehen-
sive analysis of the effects of ordinary nutrients on hepatitis C virus RNA replication
in cell culture, Antimicrob. Agents Chemother. 51 (2007) 2016–2027.
The Transparency document associated with this article can be [28] J.A. Gutierrez, K.A. Jones, R. Flores, A. Singhania, C.H. Woelk, R.T. Schooley, D.L.
found, in online version. Wyles, Vitamin D metabolites inhibit hepatitis C virus and modulate cellular
Gene expression, J. Virol. Antivir. Res. 3 (2014) http://dx.doi.org/10.4172/2324-
8955.1000129.
Acknowledgements [29] M. Gal-Tanamy, L. Bachmetov, A. Ravid, R. Koren, A. Erman, R. Tur-Kaspa, R. Zemel,
Vitamin D: an innate antiviral agent suppressing hepatitis C virus in human hepa-
The authors have no relevant conflicts of interest. JEG received tocytes, Hepatology 54 (2011) 1570–1579.
[30] K. Yuasa, A. Naganuma, K. Sato, M. Ikeda, N. Kato, H. Takagi, M. Mori, Zinc is a neg-
funding from the National Health and Medical Research Council of ative regulator of hepatitis C virus RNA replication, Liver Int. 26 (2006) 1111–1118.
Australia (AppID 1043199). [31] M.T. Kitson, C. Sarrazin, P. Toniutto, G.D. Eslick, S.K. Roberts, Vitamin D level and
sustained virologic response to interferon-based antiviral therapy in chronic hep-
atitis C: a systematic review and meta-analysis, J. Hepatol. 61 (2014) 1247–1252.
References
[32] M. Garcia-Alvarez, D. Pineda-Tenor, M.A. Jimenez-Sousa, A. Fernandez-Rodriguez,
[1] M.F. Holick, Vitamin D: evolutionary, physiological and health perspectives, Curr. M. Guzman-Fulgencio, S. Resino, Relationship of vitamin D status with advanced
Drug Targets 12 (2011). liver fibrosis and response to hepatitis C virus therapy: a meta-analysis,
[2] D.E. Prosser, G. Jones, Enzymes involved in the activation and inactivation of vita- Hepatology 60 (2014) 1541–1550.
min D, Trends Biochem. Sci. 29 (2004) 664–673. [33] D. Pineda-Tenor, M. Garcia-Alvarez, M.A. Jimenez-Sousa, A. Fernandez-Rodriguez,
[3] J.B. Cheng, M.A. Levine, N.H. Bell, D.J. Mangelsdorf, D.W. Russell, Genetic evidence S. Resino, Reply: to PMID 24975775, Hepatology 62 (2015) 1643.
that the human CYP2R1 enzyme is a key vitamin D 25-hydroxylase, Proc. Natl. [34] A. Belle, E. Gizard, G. Conroy, A. Lopez, M. Bouvier-Alias, S. Rouanet, L. Peyrin-
Acad. Sci. U. S. A. 101 (2004) 7711–7715. Biroulet, J.-M. Pawlotsky, J.-P. Bronowicki, 25-OH vitamin D level has no
[4] M.F. Holick, T.C. Chen, Vitamin D deficiency: a worldwide problem with health impact on the efficacy of antiviral therapy in naïve genotype 1 HCV-infected pa-
consequences, Am. J. Clin. Nutr. 87 (2008) 1080s–1086s. tients, United European Gastroenterol. J. (2016) http://dx.doi.org/10.1177/
[5] R. St-Arnaud, Targeted inactivation of vitamin D hydroxylases in mice, Bone 25 2050640616640157.
(1999) 127–129. [35] E. Loftfield, T.R. O'Brien, R.M. Pfeiffer, C.D. Howell, R. Horst, L. Prokunina-Olsson, S.J.
[6] T.D. Thacher, B.L. Clarke, Vitamin D insufficiency, Mayo Clin. Proc. 86 (2011) 50–60. Weinstein, D. Albanes, T.R. Morgan, N.D. Freedman, Vitamin D status and virologic
H. Elangovan et al. / Biochimica et Biophysica Acta 1863 (2017) 907–916 915

response to HCV therapy in the HALT-C and VIRAHEP-C trials, PLoS One 11 (2016), receptor triggering of a vitamin D-mediated human antimicrobial response, Sci-
e0166036. . ence 311 (2006) 1770–1773.
[36] A.C. Ross, J.E. Manson, S.A. Abrams, J.F. Aloia, P.M. Brannon, S.K. Clinton, R.A. [58] E. Zitt, H. Sprenger-Mahr, F. Knoll, U. Neyer, K. Lhotta, Vitamin D deficiency is asso-
Durazo-Arvizu, J.C. Gallagher, R.L. Gallo, G. Jones, C.S. Kovacs, S.T. Mayne, C.J. ciated with poor response to active hepatitis B immunisation in patients with
Rosen, S.A. Shapses, The 2011 report on dietary reference intakes for calcium chronic kidney disease, Vaccine 30 (2012) 931–935.
and vitamin D from the Institute of Medicine: what clinicians need to know, J. [59] R. Jhorawat, S. Jain, A. Pal, S. Nijhawan, P. Beniwal, D. Agarwal, V. Malhotra, Effect of
Clin. Endocrinol. Metab. 96 (2011) 53–58. vitamin D level on the immunogenicity to hepatitis B vaccination in dialysis pa-
[37] B. Terrier, N. Lapidus, S. Pol, L. Serfaty, V. Ratziu, T. Asselah, V. Thibault, J.-C. tients, Indian J. Gastroenterol. 35 (2016) 67–71.
Souberbielle, F. Carrat, P. Cacoub, Vitamin D in addition to peg-interferon-alpha/ri- [60] G. Targher, L. Bertolini, L. Scala, M. Cigolini, L. Zenari, G. Falezza, G. Arcaro, Associ-
bavirin in chronic hepatitis C virus infection: ANRS-HC25-VITAVIC study, World J. ations between serum 25-hydroxyvitamin D 3 concentrations and liver histology
Gastroenterol. 21 (2015) 5647–5653. in patients with non-alcoholic fatty liver disease, Nutr. Metab. Cardiovasc. Dis. 17
[38] K. Baur, J.C. Mertens, J. Schmitt, R. Iwata, B. Stieger, P. Frei, B. Seifert, H.A. (2007) 517–524.
Bischoff Ferrari, A. von Eckardstein, B. Mullhaupt, A. Geier, The vitamin D re- [61] X. Wang, W. Li, Y. Zhang, Y. Yang, G. Qin, Association between vitamin D and non-
ceptor gene bAt (CCA) haplotype impairs the response to pegylated-interfer- alcoholic fatty liver disease/non-alcoholic steatohepatitis: results from a meta-
on/ribavirin-based therapy in chronic hepatitis C patients, Antivir. Ther. 17 analysis, Int. J. Clin. Exp. Med. 8 (2015) 17221–17234.
(2012) 541–547. [62] M. Eliades, E. Spyrou, N. Agrawal, M. Lazo, F.L. Brancati, J.J. Potter, Meta-analysis:
[39] E. Garcia-Martin, J.A. Agundez, M.L. Maestro, A. Suarez, M. Vidaurreta, C. Martinez, vitamin D and non-alcoholic fatty liver disease, Aliment. Pharmacol. Ther. 38
C. Fernandez-Perez, L. Ortega, J.M. Ladero, Influence of vitamin D-related gene (2013).
polymorphisms (CYP27B and VDR) on the response to interferon/ribavirin therapy [63] G.E. Chung, D. Kim, M.-S. Kwak, J.I. Yang, J.Y. Yim, S.H. Lim, M. Itani, The serum vi-
in chronic hepatitis C, PLoS One 8 (2013), e74764. . tamin D level is inversely correlated with nonalcoholic fatty liver disease, Clin. Mol.
[40] C.-H. Hung, T.-H. Hu, S.-N. Lu, C.-H. Chen, J.-H. Wang, C.-M. Lee, Association of vita- Hepatol. 22 (2016) 146–151.
min D receptor gene polymorphisms with response to peginterferon plus ribavirin [64] F. Bril, M. Maximos, P. Portillo-Sanchez, D. Biernacki, R. Lomonaco, S. Subbarayan,
in Asian patients with chronic hepatitis C, J. Formos. Med. Assoc. 115 (2016) Relationship of vitamin D with insulin resistance and disease severity in non-alco-
278–283. holic steatohepatitis, J. Hepatol. 62 (2015).
[41] T. Arai, M. Atsukawa, A. Tsubota, C. Kondo, N. Shimada, H. Abe, N. Itokawa, A. [65] R. Anty, A. Hastier, C.M. Canivet, S. Patouraux, A.-S. Schneck, P. Ferrari-Panaia, I.
Nakagawa, T. Okubo, Y. Aizawa, K. Iwakiri, Vitamin D-related gene polymorphisms Ben-Amor, M.C. Saint-Paul, J. Gugenheim, P. Gual, A. Iannelli, A. Tran, Severe vita-
do not influence the outcome and serum vitamin D level in pegylated interferon/ min D deficiency is not associated with liver damage in morbidly obese patients,
ribavirin therapy combined with protease inhibitor for patients with genotype 1b Obes. Surg. 26 (2016) 2138–2143.
chronic hepatitis C, J. Med. Virol. 87 (2015) 1904–1912. [66] R. Diez Rodriguez, M.D. Ballesteros Pomar, A. Calleja Fernandez, S. Calleja Antolin, I.
[42] S. Abu-Mouch, Z. Fireman, J. Jarchovsky, A.-R. Zeina, N. Assy, Vitamin D supplemen- Cano Rodriguez, P. Linares Torres, F. Jorquera Plaza, J.L. Olcoz Goni, Vitamin D levels
tation improves sustained virologic response in chronic hepatitis C (genotype 1)- and bone turnover markers are not related to non-alcoholic fatty liver disease in
naïve patients, World J. Gastroenterol. 17 (2011) 5184–5190. severely obese patients, Nutr. Hosp. 30 (2014) 1256–1262.
[43] A. Nimer, A. Mouch, Vitamin D improves viral response in hepatitis C genotype 2-3 [67] Y.A. Patel, R. Henao, C.A. Moylan, C.D. Guy, D.L. Piercy, A.M. Diehl, M.F. Abdelmalek,
naive patients, World J. Gastroenterol. 18 (2012) 800–805. Vitamin D is not associated with severity in NAFLD: results of a paired clinical and
[44] A. Grasso, F. Malfatti, P. De Leo, H. Martines, P. Fabris, F. Toscanini, M. Anselmo, G. gene expression profile analysis, Am. J. Gastroenterol. (2016).
Menardo, Insulin resistance predicts rapid virological response in non-diabetic, [68] C.L. Roth, C.T. Elfers, D.P. Figlewicz, S.J. Melhorn, G.J. Morton, A. Hoofnagle, M.M.
non-cirrhotic genotype 1 HCV patients treated with peginterferon alpha-2b plus ri- Yeh, J.E. Nelson, K.V. Kowdley, Vitamin D deficiency in obese rats exacerbates non-
bavirin, J. Hepatol. 51 (2009) 984–990. alcoholic fatty liver disease and increases hepatic resistin and toll-like receptor ac-
[45] S. Yokoyama, S. Takahashi, Y. Kawakami, C.N. Hayes, H. Kohno, H. Kohno, K. Tsuji, tivation, Hepatology 55 (2012) 1103–1111.
Y. Aisaka, S. Kira, K. Yamashina, M. Nonaka, T. Moriya, M. Kitamoto, S. Aimitsu, T. [69] T. Nakano, Y.F. Cheng, C.Y. Lai, L.W. Hsu, Y.C. Chang, J.Y. Deng, Y.Z. Huang, H. Honda,
Nakanishi, H. Kawakami, K. Chayama, Effect of vitamin D supplementation on K.D. Chen, C.C. Wang, K.W. Chiu, B. Jawan, H.L. Eng, S. Goto, C.L. Chen, Impact of ar-
pegylated interferon/ribavirin therapy for chronic hepatitis C genotype 1b: a ran- tificial sunlight therapy on the progress of non-alcoholic fatty liver disease in rats, J.
domized controlled trial, J. Viral Hepat. 21 (2014) 348–356. Hepatol. 55 (2011) 415–425.
[46] J.M. Ladero, M.J. Torrejon, P. Sanchez-Pobre, A. Suarez, F. Cuenca, V. de la Orden, [70] U. Zeitz, K. Weber, D.W. Soegiarto, E. Wolf, R. Balling, R.G. Erben, Impaired insulin
M.J. Devesa, M. Rodrigo, V. Estrada, G. Lopez-Alonso, J.A. Agundez, Vitamin D defi- secretory capacity in mice lacking a functional vitamin D receptor, FASEB J. 17
ciency and vitamin D therapy in chronic hepatitis C, Ann. Hepatol. 12 (2013) (2003) 509–511.
199–204. [71] B. Maestro, S. Molero, S. Bajo, N. Davila, C. Calle, Transcriptional activation of the
[47] G. Esmat, M. El Raziky, A. Elsharkawy, D. Sabry, M. Hassany, A. Ahmed, N. Assem, human insulin receptor gene by 1,25-dihydroxyvitamin D(3), Cell Biochem.
M. El Kassas, W. Doss, Impact of vitamin D supplementation on sustained virolog- Funct. 20 (2002) 227–232.
ical response in chronic hepatitis C genotype 4 patients treated by pegylated inter- [72] K.M. Alkharfy, N.M. Al-Daghri, S.M. Yakout, T. Hussain, A.K. Mohammed, S.
feron/ribavirin, J. Interf. Cytokine Res. 35 (2015) 49–54. Krishnaswamy, Influence of vitamin D treatment on transcriptional regulation of
[48] H. Farnik, J. Bojunga, A. Berger, R. Allwinn, O. Waidmann, B. Kronenberger, O.T. insulin-sensitive genes, Metab. Syndr. Relat. Disord. 11 (2013) 283–288.
Keppler, S. Zeuzem, C. Sarrazin, C.M. Lange, Low vitamin D serum concentration [73] P. Manna, S.K. Jain, Vitamin D up-regulates glucose transporter 4 (GLUT4) translo-
is associated with high levels of hepatitis B virus replication in chronically infected cation and glucose utilization mediated by cystathionine-gamma-lyase (CSE) acti-
patients, Hepatology 58 (2013) 1270–1276. vation and H2S formation in 3T3L1 adipocytes, J. Biol. Chem. 287 (2012)
[49] N.X. Hoan, N. Khuyen, M.T. Binh, D.P. Giang, H. Van Tong, P.Q. Hoan, N.T. Trung, D.T. 42324–42332.
Anh, N.L. Toan, C.G. Meyer, P.G. Kremsner, T.P. Velavan, L.H. Song, Association of vi- [74] S. Yu, D. Bruce, M. Froicu, V. Weaver, M.T. Cantorna, Failure of T cell homing, re-
tamin D deficiency with hepatitis B virus-related liver diseases, BMC Infect. Dis. 16 duced CD4/CD8alphaalpha intraepithelial lymphocytes, and inflammation in the
(2016) 507. gut of vitamin D receptor KO mice, Proc. Natl. Acad. Sci. U. S. A. 105 (2008)
[50] X.Y. Zhao, J. Li, J.H. Wang, S. Habib, W. Wei, S.J. Sun, H.W. Strobel, J.D. Jia, Vitamin D 20834–20839.
serum level is associated with Child-Pugh score and metabolic enzyme imbalances, [75] J.E. Nelson, C.L. Roth, L.A. Wilson, K.P. Yates, B. Aouizerat, V. Morgan-Stevenson, E.
but not viral load in chronic hepatitis B patients, Medicine (Baltimore) 95 (2016), Whalen, A. Hoofnagle, M. Mason, V. Gersuk, M.M. Yeh, K.V. Kowdley, Vitamin D de-
e3926. . ficiency is associated with increased risk of non-alcoholic steatohepatitis in adults
[51] H.L. Chan, M. Elkhashab, H. Trinh, W.Y. Tak, X. Ma, W.L. Chuang, Association of with non-alcoholic fatty liver disease: possible role for MAPK and NF-kappaB? Am.
baseline vitamin D levels with clinical parameters and treatment outcomes in J. Gastroenterol. 111 (2016) 852–863.
chronic hepatitis B, J. Hepatol. 63 (2015). [76] S. Kaser, A. Moschen, A. Cayon, A. Kaser, J. Crespo, F. Pons-Romero, C.F. Ebenbichler,
[52] H.L. Chan, M. Elkhashab, H. Trinh, W.Y. Tak, X. Ma, W.L. Chuang, Y.J. Kim, E.B. J.R. Patsch, H. Tilg, Adiponectin and its receptors in non-alcoholic steatohepatitis,
Martins, L. Lin, P. Dinh, P. Charuworn, G.R. Foster, P. Marcellin, Association of base- Gut 54 (2005) 117–121.
line vitamin D levels with clinical parameters and treatment outcomes in chronic [77] J.M. Hui, A. Hodge, G.C. Farrell, J.G. Kench, A. Kriketos, J. George, Beyond insulin re-
hepatitis B, J. Hepatol. 63 (2015) 1086–1092. sistance in NASH: TNF-alpha or adiponectin? Hepatology 40 (2004) 46–54.
[53] L. Boglione, J. Cusato, A. De Nicolò, G. Cariti, G. Di Perri, A. D'Avolio, Role of [78] A. Xu, Y. Wang, H. Keshaw, L.Y. Xu, K.S.L. Lam, G.J.S. Cooper, The fat-derived hor-
CYP27B1 + 2838 promoter polymorphism in the treatment of chronic hepatitis mone adiponectin alleviates alcoholic and nonalcoholic fatty liver diseases in
B HBeAg negative with PEG-interferon, J. Viral Hepat. 22 (2015) 318–327. mice, J. Clin. Investig. 112 (2003) 91–100.
[54] Y.W. Huang, Y.T. Liao, W. Chen, C.L. Chen, J.T. Hu, C.J. Liu, M.Y. Lai, P.J. Chen, D.S. [79] L. Miele, V. Valenza, G. La Torre, M. Montalto, G. Cammarota, R. Ricci, R. Mascianà,
Chen, S.S. Yang, J.H. Kao, Vitamin D receptor gene polymorphisms and distinct clin- A. Forgione, M.L. Gabrieli, G. Perotti, F.M. Vecchio, G. Rapaccini, G. Gasbarrini, C.P.
ical phenotypes of hepatitis B carriers in Taiwan, Genes Immun. 11 (2010) 87–93. Day, A. Grieco, Increased intestinal permeability and tight junction alterations in
[55] J.H. Li, H.Q. Li, Z. Li, Y. Liu, J.R. Gao, X.J. Zeng, C.Y. Gou, X.L. Zhu, X.H. Guo, L. Pan, H. Li, nonalcoholic fatty liver disease, Hepatology 49 (2009) 1877–1887.
Association of Taq I T/C and Fok I C/T polymorphisms of vitamin D receptor gene [80] K.L. Donnelly, C.I. Smith, S.J. Schwarzenberg, J. Jessurun, M.D. Boldt, E.J. Parks,
with outcome of hepatitis B virus infection, Zhonghua Yi Xue Za Zhi 86 (2006) Sources of fatty acids stored in liver and secreted via lipoproteins in patients
1952–1956. with nonalcoholic fatty liver disease, J. Clin. Invest. 115 (2005) 1343–1351.
[56] R. Bellamy, C. Ruwende, T. Corrah, K.P.W.J. McAdam, M. Thursz, H.C. Whittle, A.V.S. [81] C. Ning, L. Liu, G. Lv, Y. Yang, Y. Zhang, R. Yu, Y. Wang, J. Zhu, Lipid metabolism and
Hill, Tuberculosis and chronic hepatitis B virus infection in Africans and variation in inflammation modulated by Vitamin D in liver of diabetic rats, Lipids Health Dis. 14
the vitamin D receptor gene, J. Infect. Dis. 179 (1999) 721–724. (2015) 1–9.
[57] P.T. Liu, S. Stenger, H. Li, L. Wenzel, B.H. Tan, S.R. Krutzik, M.T. Ochoa, J. Schauber, K. [82] I. Barchetta, M. Del Ben, F. Angelico, M. Di Martino, A. Fraioli, G. La Torre, R. Saulle, L.
Wu, C. Meinken, D.L. Kamen, M. Wagner, R. Bals, A. Steinmeyer, U. Zugel, R.L. Gallo, Perri, S. Morini, C. Tiberti, L. Bertoccini, F.A. Cimini, F. Panimolle, C. Catalano, M.G.
D. Eisenberg, M. Hewison, B.W. Hollis, J.S. Adams, B.R. Bloom, R.L. Modlin, Toll-like Baroni, M.G. Cavallo, No effects of oral vitamin D supplementation on non-
916 H. Elangovan et al. / Biochimica et Biophysica Acta 1863 (2017) 907–916

alcoholic fatty liver disease in patients with type 2 diabetes: a randomized, double- [102] S. Pilz, C. Putz-Bankuti, M. Gaksch, W. Spindelboeck, M. Haselberger, F. Rainer, A.
blind, placebo-controlled trial, BMC Med. 14 (2016) 92. Posch, P. Kreuzer, T. Stojakovic, V. Stadlbauer, B. Obermayer-Pietsch, R. Stauber, Ef-
[83] M.T. Kitson, A. Pham, A. Gordon, W. Kemp, S.K. Roberts, High-dose vitamin D sup- fects of vitamin D supplementation on serum 25-hydroxyvitamin D concentrations
plementation and liver histology in NASH, Gut 65 (2016). in cirrhotic patients: a randomized controlled trial, Nutrients 8 (2016) 278.
[84] N. Sharifi, R. Amani, E. Hajiani, B. Cheraghian, Does vitamin D improve liver en- [103] S. Ruiz-Gaspa, N. Guanabens, A. Enjuanes, P. Peris, A. Martinez-Ferrer, M.J. de
zymes, oxidative stress, and inflammatory biomarkers in adults with non-alcoholic Osaba, B. Gonzalez, L. Alvarez, A. Monegal, A. Combalia, A. Pares, Lithocholic acid
fatty liver disease? A randomized clinical trial, Endocrine 47 (2014) 70–80. downregulates vitamin D effects in human osteoblasts, Eur. J. Clin. Investig. 40
[85] L. Fan, X. Tu, Y. Zhu, L. Zhou, T. Pfeiffer, R. Feltens, W. Stoecker, R. Zhong, Genetic (2010) 25–34.
association of vitamin D receptor polymorphisms with autoimmune hepatitis [104] A. Duran, E.D. Hernandez, M. Reina-Campos, E.A. Castilla, S. Subramaniam, S.
and primary biliary cirrhosis in the Chinese, J. Gastroenterol. Hepatol. 20 (2005) Raghunandan, L.R. Roberts, T. Kisseleva, M. Karin, M.T. Diaz-Meco, J. Moscat, p62/
249–255. SQSTM1 by binding to vitamin D receptor inhibits hepatic stellate cell activity, fi-
[86] B. Halmos, F. Szalay, T. Cserniczky, E. Nemesanszky, P. Lakatos, S. Barlage, G. brosis, and liver cancer, Cancer Cell 30 (2016) 595–609.
Schmitz, L. Romics, A. Csaszar, Association of primary biliary cirrhosis with vitamin [105] K.C. Chiang, K.S. Persons, N.W. Istfan, M.F. Holick, T.C. Chen, Fish oil enhances the
D receptor BsmI genotype polymorphism in a Hungarian population, Dig. Dis. Sci. antiproliferative effect of 1alpha,25-dihydroxyvitamin D3 on liver cancer cells, An-
45 (2000) 1091–1095. ticancer Res. 29 (2009) 3591–3596.
[87] A. Vogel, C.P. Strassburg, M.P. Manns, Genetic association of vitamin D receptor [106] Z. Ghous, J. Akhter, M.H. Pourgholami, D.L. Morris, Inhibition of hepatocellular can-
polymorphisms with primary biliary cirrhosis and autoimmune hepatitis, cer by EB1089: in vitro and in vive study, Anticancer Res. 28 (2008) 3757–3761.
Hepatology 35 (2002) 126–131. [107] J. Akhter, Y. Lu, I. Finlay, M.H. Pourgholami, D.L. Morris, 1Alpha,25-
[88] L.P. Lakatos, E. Bajnok, D. Hegedus, T. Toth, P. Lakatos, F. Szalay, Vitamin D receptor, dihydroxyvitamin D3 and its analogues, EB1089 and CB1093, profoundly inhibit
oestrogen receptor-alpha gene and interleukin-1 receptor antagonist gene poly- the in vitro proliferation of the human hepatoblastoma cell line HepG2, ANZ J.
morphisms in Hungarian patients with primary biliary cirrhosis, Eur. J. Surg. 71 (2001) 414–417.
Gastroenterol. Hepatol. 14 (2002) 733–740. [108] D. Sahpazidou, P. Stravoravdi, T. Toliou, G. Geromichalos, G. Zafiriou, K. Natsis, P.
[89] A. Tanaka, S. Nezu, S. Uegaki, K. Kikuchi, A. Shibuya, H. Miyakawa, S.-i. Takahashi, I. Gigis, Significant experimental decrease of the hepatocellular carcinoma incidence
Bianchi, P. Zermiani, M. Podda, H. Ohira, P. Invernizzi, H. Takikawa, Vitamin D re- in C3H/Sy mice after long-term administration of EB1089, a vitamin D analogue,
ceptor polymorphisms are associated with increased susceptibility to primary bil- Oncol. Res. 13 (2003) 261–268.
iary cirrhosis in Japanese and Italian populations, J. Hepatol. 50 (2009) 1202–1209. [109] C. Pivonello, D.P. Provvisiero, M. Negri, G. Di Gennaro, C. De Angelis, G. Galdiero, C.
[90] #x. Kempi, A. ska-Podhorecka, E. Wunsch, T. Jarowicz, J. Raszeja-Wyszomirska, B. Simeoli, M.C. De Martino, A. Colao, R. Pivonello, Potential role of vitamin D in re-
Loniewska, M. Kaczmarczyk, M. Milkiewicz, #x. Gorzata, P. Milkiewicz, Vitamin D storing sensitivity to mTOR inhibitors in hepatocellular carcinoma (HCC):
receptor polymorphisms predispose to primary biliary cirrhosis and severity of 1,25(OH)vitamin D (VITD) reverts everolimus (EVE) resistance in a HCC cell line,
the disease in polish population, Gastroenterol. Res. Pract. 2012 (2012) 8. Endocr. Abstr. 41 (2016).
[91] G.Y. Guo, Y.Q. Shi, L. Wang, X. Ren, Z.Y. Han, C.C. Guo, L.N. Cui, J.B. Wang, J. Zhu, N. [110] J.A. Mertz, A.R. Conery, B.M. Bryant, P. Sandy, S. Balasubramanian, D.A. Mele, L.
Wang, J. Zhang, Y. Cai, Y. Han, X.M. Zhou, D.M. Fan, Serum vitamin D level is asso- Bergeron, R.J. Sims 3rd, Targeting MYC dependence in cancer by inhibiting BET
ciated with disease severity and response to ursodeoxycholic acid in primary bili- bromodomains, Proc. Natl. Acad. Sci. U. S. A. 108 (2011) 16669–16674.
ary cirrhosis, Aliment. Pharmacol. Ther. 42 (2015) 221–230. [111] J. Chen, L.H. Katz, N.M. Muñoz, S. Gu, J.-h. Shin, W.S. Jogunoori, M.-H. Lee, M.D.
[92] N. Agmon-Levin, R. Kopilov, C. Selmi, U. Nussinovitch, M. Sánchez-Castañón, M. Belkin, S.-B. Kim, J.C. White, J. Andricovich, A. Tzatsos, S. Li, S.S. Kim, K. Shetty, B.
López-Hoyos, H. Amital, S. Kivity, E.M. Gershwin, Y. Shoenfeld, Vitamin D in prima- Mishra, A. Rashid, J.-S. Lee, L. Mishra, Vitamin D deficiency promotes liver tumor
ry biliary cirrhosis, a plausible marker of advanced disease, Immunol. Res. 61 growth in transforming growth factor-β/Smad3-deficient mice through Wnt and
(2015) 141–146. toll-like receptor 7 pathway modulation, Sci. Rep. 6 (2016) 30217.
[93] E. Liaskou, L.E. Jeffery, P.J. Trivedi, G.M. Reynolds, S. Suresh, T. Bruns, D.H. Adams, [112] C. ETHIER, R. KESTEKIAN, C. BEAULIEU, C. DUBE, J. HAVRANKOVA, M. GASCON-
D.M. Sansom, G.M. Hirschfield, Loss of CD28 expression by liver-infiltrating T BARRE, Vitamin D depletion retards the normal regeneration process after partial
cells contributes to pathogenesis of primary sclerosing cholangitis, Gastroenterol- hepatectomy in the rat, Endocrinology 126 (1990) 2947–2959.
ogy 147 (2014) 221–232, e227. . [113] M.H. Sherman, R.T. Yu, D.D. Engle, N. Ding, A.R. Atkins, H. Tiriac, E.A. Collisson, F.
[94] C.A. Redaelli, M. Wagner, Y.H. Tien, L. Mazzucchelli, P.F. Stahel, M.K. Schilling, J.F. Connor, T. Van Dyke, S. Kozlov, P. Martin, T.W. Tseng, D.W. Dawson, T.R.
Dufour, 1 alpha,25-dihydroxycholecalciferol reduces rejection and improves sur- Donahue, A. Masamune, T. Shimosegawa, M.V. Apte, J.S. Wilson, B. Ng, S.L. Lau,
vival in rat liver allografts, Hepatology 34 (2001) 926–934. J.E. Gunton, G.M. Wahl, T. Hunter, J.A. Drebin, P.J. O'Dwyer, C. Liddle, D.A.
[95] T. Xing, G. Qiu, L. Zhong, L. Ling, L. Huang, Z. Peng, Calcitriol reduces the occurrence Tuveson, M. Downes, R.M. Evans, Vitamin D receptor-mediated stromal
of acute cellular rejection of liver transplants: a prospective controlled study, Die reprogramming suppresses pancreatitis and enhances pancreatic cancer therapy,
Pharmazie - Int. J. Pharm. Sci. 68 (2013) 821–826. Cell 159 (2014) 80–93.
[96] E. Falleti, D. Bitetto, C. Fabris, S. Cmet, E. Fornasiere, A. Cussigh, E. Fontanini, C. [114] D.L. Morris, J.L. Jourdan, I. Finlay, T. Gruenberger, M.P. The, M.H. Pourgholami, He-
Avellini, G. Barbina, E. Ceriani, M. Pirisi, P. Toniutto, Association between vitamin patic intra-arterial injection of 1,25-dihydroxyvitamin D3 in lipiodol: pilot study in
D receptor genetic polymorphisms and acute cellular rejection in liver- patients with hepatocellular carcinoma, Int. J. Oncol. 21 (2002) 901–906.
transplanted patients, Transpl. Int. 25 (2012) 314–322. [115] K. Dalhoff, J. Dancey, L. Astrup, T. Skovsgaard, K.J. Hamberg, F.J. Lofts, O. Rosmorduc,
[97] C. Andraos, G. Koorsen, J.C. Knight, L. Bornman, Vitamin D receptor gene methyla- S. Erlinger, J. Bach Hansen, W.P. Steward, T. Skov, F. Burcharth, T.R.J. Evans, A phase
tion is associated with ethnicity, tuberculosis, and TaqI polymorphism, Hum. II study of the vitamin D analogue Seocalcitol in patients with inoperable hepato-
Immunol. 72 (2011) 262–268. cellular carcinoma, Br. J. Cancer 89 (2003) 252–257.
[98] A.G. Uitterlinden, Y. Fang, J.B. Van Meurs, H.A. Pols, J.P. Van Leeuwen, Genetics and [116] A. Umemura, F. He, K. Taniguchi, H. Nakagawa, S. Yamachika, J. Font-Burgada, Z.
biology of vitamin D receptor polymorphisms, Gene 338 (2004) 143–156. Zhong, S. Subramaniam, S. Raghunandan, A. Duran, J.F. Linares, M. Reina-Campos,
[99] S. Abramovitch, L. Dahan-Bachar, E. Sharvit, Y. Weisman, A.B. Tov, E. Brazowski, Vi- S. Umemura, M.A. Valasek, E. Seki, K. Yamaguchi, K. Koike, Y. Itoh, M.T. Diaz-
tamin D inhibits proliferation and profibrotic marker expression in hepatic stellate Meco, J. Moscat, M. Karin, p62, upregulated during preneoplasia, induces hepato-
cells and decreases thioacetamide-induced liver fibrosis in rats, Gut 60 (2011). cellular carcinogenesis by maintaining survival of stressed HCC-initiating cells,
[100] K. Hochrath, C.S. Stokes, J. Geisel, M.J. Pollheimer, P. Fickert, S. Dooley, F. Lammert, Cancer Cell 29 (2016) 935–948.
Vitamin D modulates biliary fibrosis in ABCB4-deficient mice, Hepatol. Int. 8 [117] A.A. Seif, D.M. Abdelwahed, Vitamin D ameliorates hepatic ischemic/reperfusion
(2014) 443–452. injury in rats, J. Physiol. Biochem. 70 (2014) 659–666.
[101] S. Abramovitch, E. Sharvit, Y. Weisman, A. Bentov, E. Brazowski, G. Cohen, O. [118] D. Kim, M. Kim, Y. Chang, J. Kim, J. Lee, Abstracts of the 37th ESPEN Congress,
Volovelsky, S. Reif, Vitamin D inhibits development of liver fibrosis in animal Lisbon, Portugal, 5–8 September 2015SUN-PP010: the vitamin D analogue
model but cannot ameliorate established cirrhosis, Am. J. Physiol. Gastrointest. paricalcitol attenuates hepatic ischemia/reperfusion injury by modulation of
Liver Physiol. (2014). Tlr4-mediated signaling in rats, Clin. Nutr. 34 (2015) S26–S27.

You might also like