ArticlePDF AvailableLiterature Review

Genomic or Non-Genomic? A Question about the Pleiotropic Roles of Vitamin D in Inflammatory-Based Diseases

MDPI
Nutrients
Authors:

Abstract

Vitamin D (vit D) is widely known for its role in calcium metabolism and its importance for the bone system. However, various studies have revealed a myriad of extra-skeletal functions, including cell differentiation and proliferation, antibacterial, antioxidant, immunomodulatory, and anti-inflammatory properties in various cells and tissues. Vit D mediates its function via regulation of gene expression by binding to its receptor (VDR) which is expressed in almost all cells within the body. This review summarizes the pleiotropic effects of vit D, emphasizing its anti-inflammatory effect on different organ systems. It also provides a comprehensive overview of the genetic and epigenetic effects of vit D and VDR on the expression of genes pertaining to immunity and anti-inflammation. We speculate that in the context of inflammation, vit D and its receptor VDR might fulfill their roles as gene regulators through not only direct gene regulation but also through epigenetic mechanisms.
Citation: Holick, M.F.; Mazzei, L.;
García Menéndez, S.; Martín
Giménez, V.M.; Al Anouti, F.;
Manucha, W. Genomic or
Non-Genomic? A Question about the
Pleiotropic Roles of Vitamin D in
Inflammatory-Based Diseases.
Nutrients 2023,15, 767. https://
doi.org/10.3390/nu15030767
Academic Editor: Carsten Carlberg
Received: 6 December 2022
Revised: 10 January 2023
Accepted: 18 January 2023
Published: 2 February 2023
Copyright: © 2023 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
nutrients
Review
Genomic or Non-Genomic? A Question about the Pleiotropic
Roles of Vitamin D in Inflammatory-Based Diseases
Michael F. Holick 1,* , Luciana Mazzei 2,3, Sebastián García Menéndez 2,3 , Virna Margarita Martín Giménez 4,
Fatme Al Anouti 5and Walter Manucha 3,6,*
1Section on Endocrinology, Diabetes, Nutrition & Weight Management, Department of Medicine, School of
Medicine, Boston University, Boston, MA 02118, USA
2Instituto de Bioquímica y Biotecnología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo,
Mendoza 5500, Argentina
3Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones
Científicas y Tecnológicas (CONICET), Mendoza 5500, Argentina
4Instituto de Investigaciones en Ciencias Químicas, Facultad de Ciencias Químicas y Tecnológicas,
Universidad Católica de Cuyo, San Juan 5400, Argentina
5Department of Health Sciences, Zayed University, Abu Dhabi P.O. Box 144534, United Arab Emirates
6Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de
Cuyo, Mendoza 5500, Argentina
*Correspondence: mfholick@bu.edu (M.F.H.); wmanucha@fcm.uncu.edu.ar (W.M.);
Tel.: +54-261-4494116 (W.M.)
Abstract:
Vitamin D (vit D) is widely known for its role in calcium metabolism and its importance
for the bone system. However, various studies have revealed a myriad of extra-skeletal functions,
including cell differentiation and proliferation, antibacterial, antioxidant, immunomodulatory, and
anti-inflammatory properties in various cells and tissues. Vit D mediates its function via regulation of
gene expression by binding to its receptor (VDR) which is expressed in almost all cells within the body.
This review summarizes the pleiotropic effects of vit D, emphasizing its anti-inflammatory effect on
different organ systems. It also provides a comprehensive overview of the genetic and epigenetic
effects of vit D and VDR on the expression of genes pertaining to immunity and anti-inflammation.
We speculate that in the context of inflammation, vit D and its receptor VDR might fulfill their roles
as gene regulators through not only direct gene regulation but also through epigenetic mechanisms.
Keywords: vitamin D; genetic; genomic; epigenetic; immunity; inflammation
1. Introduction
Vitamin D (vit D) is a fat-soluble vitamin that can be found in two different chemical
structures: cholecalciferol (vit D3) or ergocalciferol (vit D2) [
1
]. The exposure to ultraviolet
B rays, present in sunlight, is the most effective natural source of vit D. It also can be
obtained by the consumption of some fatty fish (tuna, salmon, and mackerel), sun-exposed
mushrooms, beef liver, cheese, egg yolks, fortified foods, or even by supplements [2].
The active form of the vit D (D represents either and/or vitamin D2 and vitamin D3)
is 1,25-dihydroxyvitamin D (1,25(OH)2D) To become 1,25(OH)2D, first vit D is transported
to the liver, where it is modified to 25-hydroxyvitamin D (25(OH)D) by CYP2R1, which
is a 25-hydroxylase enzyme. Later, in the kidneys, 25(OH)D undergoes an additional
hydroxylation by the enzyme 25 hydroxyvitamin D-1
α
-hydroxylase (CYP27B1), converting
it to 1,25(OH)2D [3,4].
There is a lot of controversy about the optimal levels of 25(OH)D which is the measure
of a person’s vitamin D status [
5
]. Most agreements indicate vit D deficiency is defined
at values below 20 ng/mL, insufficiency between 21–29 ng/mL and sufficiency at values
> 30 ng/mL, with the range between 40–60 ng/mL being preferred, and vit D intoxication
at values above 150 ng/mL as recommended by the Endocrine Society Practice Guidelines
Nutrients 2023,15, 767. https://doi.org/10.3390/nu15030767 https://www.mdpi.com/journal/nutrients
Nutrients 2023,15, 767 2 of 13
on Vitamin D [
6
]. However, according to the Institute of Medicine (IOM), for the majority of
the population, a minimum 25(OH)D serum level of 20 ng/mL (50 nmol/L) is considered
adequate with limited sun exposure. Meanwhile, the risk of vit D deficiency is considered
significant when the 25(OH)D serum concentrations are below 12 ng/mL (30 nmol/L) [
7
].
Such thresholds of serum 25(OH)D concentrations were established for maximum bone
health and higher thresholds have been recommended for optimal overall health because of
the strong association between vit D deficiency and cardiovascular diseases, certain types
of cancer, type 2 diabetes mellitus, hypertension, metabolic syndrome, infectious diseases,
autism, depression, autoimmune diseases, and others [8].
In this context, given that the inflammatory process is directly or indirectly involved
in the main underlying mechanisms of most of the pathologies affecting current societies
(such as those previously mentioned), the present review aims to summarize the pleiotropic
effects of vit D, emphasizing its anti-inflammatory impact on different organ systems.
Likewise, this review aims to provide a comprehensive overview of the genomic and
non-genomic effects of vit D and vit D receptor (VDR) related to immunity and anti-
inflammation. Deepening this knowledge would help develop new therapeutic alternatives
for many diseases that constitute important causes of morbidity and mortality worldwide.
2. Pleiotropic Effects of Vitamin D in Inflammatory-Based Diseases
Numerous pleiotropic effects have been reported for vit D since its discovery a century
ago. Today it is well known that in addition to its pivotal role in calcium homeostasis
and bone metabolism, vit D has antibacterial, anti-proliferative, immunomodulatory, and
anti-inflammatory actions, among other beneficial properties [9,10].
The nuclear VDR mediates two types of actions of 1,25(OH)2D:
-Non-genomic pathway: Ligand binding to the cytosolic population of VDRs trig-
gers multiple intracellular signaling pathways or cascades, leading to immediate gene
transcription-independent responses in cells. These pathways may also contribute to
ultimately modulating the expression of target genes in the nucleus [2].
-Genomic pathway: The retinoic acid receptor (RXR) forms a heterodimer with the
VDR bound to 1,25(OH)2D. The heterodimer translocates to the cell nucleus and binds to
the vit D response element (VDRE) in the promoter regions of target genes, consequently
regulating nuclear transcription. The binding of the ligand to VDR modifies the conforma-
tion of its LBD region, usually promoting the release of transcriptional co-repressors and the
recruitment of co-activators and enzymes (histone acetylases) that modify the structure of
chromatin, facilitating the transcriptional activation of genes. In other cases, the effect could
be transcriptional repression; however, the exact mechanisms of such down-regulation are
not well known [2].
VDR is expressed in the skin, parathyroid glands, adipocytes, small intestine, colon,
and in other tissues [
11
,
12
]. Moreover, the VDRE is found in numerous genes, explaining
the mechanisms associated with vit D, such as immune functions, intestinal barrier function,
cell proliferation, gut microbiota modulation, and autophagy. Vit D immunomodulatory
effects particularly have been widely investigated by several researchers [
13
15
]. Such
effects were attributed to a direct relation to antigen-presenting cells and T-cells functions.
Moreover, the lack of 1,25(OH)2D harms regulatory T-cells differentiation and weakens its
functions, which may trigger autoimmune diseases [16,17].
In addition to its important anti-inflammatory role, vit D can exert protective effects against
reactive oxygen species (ROS) and nitric oxide and may prevent oxidative damages [
10
]. It
also reduces glutathione (GSH) synthesis, which is essential to glutathione peroxidase (GPX)
activity, through the regulation of the gamma-glutamyl transpeptidase (g-GT) expression. Vit
D cans also increase GSH formation by increasing glucose-6-phosphate dehydrogenase (G6PD),
glutamate-cysteine ligase (CGL), and glutathione reductase activity [18].
Furthermore, vit D plays a role in antioxidants synthesis by regulating Klotho and
Nrf2 (Nuclear factor erythroid 2–related factor 2) expression, both of which are essential to
ROS signaling pathway function [19].
Nutrients 2023,15, 767 3 of 13
Vit D is involved in the regulation of muscle development and contractility through
genomic actions, and stimulating the proliferation of muscle cells and their differentiation
through the transcription of genes that express an increase in cellular DNA synthesis, followed
by the induction of muscle proteins, specifically calcium and myosin binding proteins [
20
].
Vit D also exerts non-genomic actions by interacting with the specific muscle cell membrane
receptor, which leads to the stimulation of adenyl cyclase and phospholipases C, D, and A2,
and the activation of intracellular signaling pathways, such as MAPK (Mitogen-activated
protein kinase) cascade, ultimately enhancing cell division [
21
]. In a recently published study
by Max et al. [
22
], the authors observed that mice born from mothers with vit D deficiency had
smaller muscle cells than mice whose mothers had adequate levels. It has long been observed
that vit D deficiency leads to a myopathy characterized by proximal muscle weakness and
atrophy, and the presence of VDR in skeletal muscle tissue has been supported by several
studies which documented a decline in receptors with age [23].
2.1. The Role of Vitamin D in Inflammation at the Cardiovascular Level
Vit D deficiency is associated with an increase in serum levels of pro-inflammatory me-
diators, including IL-6 and tumor necrosis factor-alpha (TNF-
α
), which are related to both
the development and progression of some vascular inflammatory pathologies [
14
,
15
,
24
,
25
].
In addition, a study carried out on obese patients revealed that reduced serum 25(OH)D
concentrations were usually related to increased levels of other biomarkers of vascular
inflammation such as high-sensitivity C reactive protein (hCRP) and fibrinogen [
26
]. Sim-
ilar conclusions were informed for severely obese children [
27
,
28
]. The 25(OH)D levels
below 20 ng/mL were associated with increased markers of oxidative/nitrosative stress,
inflammation and endothelial activation, all of them indicators of cardiovascular risk.
Moreover, in obese people, hyperleptidemia is usually observed. This disorder provokes
vascular inflammation [
29
]. The 1,25(OH)2D3 pretreatment of cultured human umbilical
vein-derived endothelial cells exposed to high concentrations of leptin with 1,25(OH)2D3
prevented the rise in the expression of vascular pro-inflammatory mediators caused by
leptin, including CCL2, VCAM-1, and transforming growth factor
β
(TGF-
β
) [
30
]. Further-
more, Oma et al. [
31
] observed a higher presence of mononuclear cell infiltrates in the aortic
adventitia of patients with coronary artery disease and vit D deficiency as compared with
those with suiTable 25(OH)D levels. The adhesion of monocytes to endothelial cells repre-
sents an early stage in atherosclerosis development, thus, vit D supplementation would be
a helpful tool in the prevention and treatment of atherosclerosis and other vascular inflam-
matory diseases [
32
]. Furthermore, it has also been suggested that vit D deficiency and the
down-regulation of its receptor could be involved in aggravating vascular inflammation
in pregnant women during preeclampsia (pregnancy hypertension). Interestingly, vit D
supplementation reversed the inflammatory process in these patients [
33
]. These findings
along with the significant number of VDR located in vascular smooth muscle cells (VSMCs)
and endothelial cells indicates a crucial role of this endogenous compound in the regulation
of inflammation, especially at the vascular level [34].
The location of VDR in VSMCs is of particular interest since these cells represent
the majority of cell population in the blood vessel walls’ levels and they have a vital role
in the advance of vascular inflammatory disease. New technologies have shown that
VSMCs may switch their contractile phenotype to a macrophage-like phenotype, proving
the plasticity of these cells. Thus, there is interplay among VSMCs, immune cells, and
endothelial cells during the convoluted process of vascular inflammation where VSMCs
can control, interact with, and influence the behavior of other cellular components of
the blood vessel wall [
35
]. In this context, a study carried out using an
in vitro
model
of endothelial inflammation (primary cultured human umbilical vein endothelial cells
exposed to TNF-
α
) showed that the treatment with paricalcitol (a vit D analog) inhibited
the increased expression of the intercellular adhesion molecule-1 (ICAM-1), vascular cell
adhesion molecule-1 (VCAM-1), and fractalkine (a chemoattractant cytokine) in these
cells [
36
]. Furthermore, in a clinical model of vascular inflammation (patients with an
Nutrients 2023,15, 767 4 of 13
abdominal aneurysm), paricalcitol reduced the CD4+ T-helper and the T-cell (CD3+) content
in aneurysm wall samples from these patients. Paricalcitol also prevented the rise in the
levels of pro-inflammatory cytokines (IL-1
β
, IL-6, IL-8, and TNF-
α
) in cultured human
aortic smooth muscle cells exposed to elevated concentrations of phosphate, indicating that
active vit D derivatives have anti-inflammatory effects at the vascular level [37].
Likewise, VDRs also modulate the RAAS, inflammation, and fibrosis. Thus, their
activation counteracts myocardial hypertrophy and hypertension [
38
]. In this regard, it
is proposed that the activation of VDRs associated with Hsp70 (as a chaperone protein)
could favor physiological cardiac remodeling after a myocardial infarction and reduce
progression to heart failure [39].
In SHR rats, treatment with vit D analogs ameliorates left ventricular hypertrophy and
improves left ventricular diastolic measures. Mizobuchi et al. [
40
] showed that combined
therapy with enalapril and paricalcitol significantly decreased proteinuria, glomerulo-
sclerotic index, and tubulointerstitial volume in uremic rats [41].
In addition, experimental studies suggest that active vit D analogues at low doses
ameliorate myocardial renin overexpression and lower blood pressure, protect against
aortic calcification and prevent cardiac/vascular remodeling [42].
Moreover, our previous work [
38
] in rats with a ureteral obstruction showed a re-
duction in myocardial VDR expression, and this fact, might be related to myocardial
remodeling associated with an increase in arrhythmogenesis. Important to note, that
paricalcitol protects against these changes by restoring myocardial VDR levels [38].
For its part, vit D deficiency at the cellular level produces a higher oxidative stress,
inflammatory markers, and mitochondrial damage. Serum 25(OH)D concentrations below
25 ng/mL were related to an increase in vascular tone mediated by smooth muscle contraction,
either through direct effects on vascular smooth muscle cells, up-regulation of the RAAS,
and/or through modulation of calcium metabolism with secondary hyperparathyroidism;
which predisposes patients to develop hypertrophy of the left ventricle and of the vascular wall,
causing hypertension. The VDR plays a role at the mitochondrial level and the regulation
of the respiratory chain, which would influence arterial remodeling, since its activation
would reduce oxidative damage and preserve cell life. Such data implicates that maintaining
adequate levels of vit D is important for protection against cardiovascular disease [42].
Several studies assessed the effect of calcitriol administration as an anti-inflammatory
agent. On an apoE (
/
) mouse model of abdominal aneurysm induced by angiotensin-II
(Ang-II) infusion, the oral treatment with calcitriol reduced the aneurysm formation and
all altered parameters observed during abdominal aneurysm were decreased, including
macrophages infiltration, expression of vascular endothelial growth factor (VEGF), angio-
genesis, monocyte chemoattractant protein 1 (CCL2), CCL5, and CXCL1 chemokines and
synthesis of matrix metalloproteinase-2 and 9 (MMP-2 and 9) [
43
]. In an
in vitro
model of
human endothelial cells, the treatment caused inhibition of leukocyte-endothelial cell inter-
actions induced by Ang-II, morphogenesis, and synthesis of endothelial pro-inflammatory
and angiogenic chemokines mediated by VDRs [43,44].
In addition, vit D administration decreases the expression of pro-inflammatory and
proatherogenic cytokines such as IL-2 and interferon-gamma (IFN-
7
), which are responsible
for the T-helper-1 cells activation and vascular inflammation [
45
]. Accordingly, clinical
studies showed that vit D supplementation was able to reduce central blood pressure
parameters in individuals with vit D deficiency, but not in individuals with adequate
vit D status. It was also reported to improve arterial stiffness in overweight African
Americans patients with vit D deficiency and to improve microvascular responses in African
Americans, mitigating or preventing the development of cardiovascular dysfunction in this
population [46].
2.2. Anti-Inflammatory Role of Vitamin D in the Gastrointestinal System
Vit D helps the intestine absorb calcium and phosphate and its deficiency is correlated
with increased mucosal inflammation, leading to inflammatory bowel disease (IBD) [
2
].
Nutrients 2023,15, 767 5 of 13
However, administration of the vit D at a dose that raises and maintains a 25(OH)D of
30 ng/mL, may have the ability to reduce the disease. Moreover, VDR has been linked
to the gut microbiota and its metabolites and its expression is down-regulated in Crohn’s
disease (CD) and ulcerative colitis [2].
VDR is regulated by miRNAs, which are a class of small non-coding RNA (17–22 nu-
cleotides) that regulates gene expression post-transcriptionally. It also inhibits transcription
of ZO-1, claudin-5, and occludin genes and increases the tight junction protein claudin-2
which enhance intestinal permeability. Thus, lacking intestinal epithelial VDR regulation in
inflamed intestine leads to hyperfunction of Claudin-2 and exaggerates the inflammatory
responses in the intestines [47].
2.3. Vitamin D Role in Renal Protection
Several animal models have suggested a role for active vit D in albuminuria and
kidney fibrosis. In fact, vit D analogues are able to affect blood pressure, proteinuria, and
inflammation. A study carried out on VDR knockout mice reveals a rise in renin consequent
to loss of normal suppression of the renin-angiotensin aldosterone system (RAAS) by vit
D [
48
]. Thus, RAAS have been shown to play an important role in the progression of
chronic kidney disease (CKD) and low levels of 25(OH)D and 1,25 (OH)2D were correlated
to be predictors of disease progression and death in patients with CKD and End Stage
Renal Disease [49].
In this regard, in previous investigations, the expression of VDR and genes associated
with nephrogenesis in spontaneous hypertension rats (SHR) from week 0 to 8 of life were
analyzed. Hypertension in these rats is known to develop at about 6 weeks of age [
50
]. We
observed a decrease in the expression of the nephrogenic gene, wt-1, and VDR by week
4, before the establishment of arterial hypertension, suggesting that the alteration in the
kidney occurs previous to the increase in blood pressure [51].
Moreover, our group has found in adult SHR rats that the induction of VDR modulates
an increase in Hsp70 levels, with a decrease in the angiotensin II receptor, type 1 (AT1)
expression, providing renal protection [
41
]. Of interest, Hsp70 effects on the VDR may also
accentuate repressive anti-inflammatory signaling [52].
2.4. The Role of Vitamin D in the Nervous System
As early as the early 1980s, attempts were made to find a relationship between the
nervous system and vit D, starting with the question of whether vit D was able to cross the
blood-brain barrier [
53
]. Today the questions revolve around the relationship of vit D status
and its interaction with antioxidant mechanisms, complex immunomodulatory systems,
and neurotrophic factors among others [
19
,
54
]. As a result, vit D has a key part in the
process of neuro-inflammation, cognitive decline, and neurodegeneration [
55
]. In line with
this, it has recently been reported that vit D has a neuroprotective function in aging cognitive
decline [
56
]. This is one of the reasons why many human dietary supplements include vit D
alone or in combination with other compounds that have antioxidant effects. A closer look
into the different cell types of the nervous system, VDR are found in neurons, astrocytes, and
microglia in the central nervous system [
54
]. Furthermore, using the 25-hydroxyvitamin
D-1alpha-hydroxylase (CYP27B1) and 25-hydroxyvitamin D-24-hydroxylase (CYP24A1)
enzymes, some of these cells may synthesize and catabolize 1,25(OH)2D. The unequal
distribution of 1,25(OH)2D and 25(OH) D in different parts of the brain shows that vit D and
its metabolism in the CNS might either function in a paracrine or autocrine manner [57].
Many studies have discovered that appropriate amounts of vit D reduce oxidative
stress and brain inflammation, resulting in diverse neuroprotective benefits [
58
]. The VDR
is also implicated in the neuroprotective effects of neurosteroids. Furthermore, calcitriol has
been shown to stimulate VDR expression, down-regulate NOX2, and inhibit cellular death
rate [
59
], suggesting that VDR-activated ERK1/2 activation may contribute to neuronal
apoptosis prevention [
60
]. On the other hand, findings show that vit D shortage causes
Nutrients 2023,15, 767 6 of 13
significant changes in microglia, implying that these cells may play a role in the sensory
dysfunctions associated with hypovitaminosis D [61].
The human brain produces 1,25(OH)2D3, which affects a variety of brain areas, in-
cluding the prefrontal cortex, hippocampus, cingulate gyrus, thalamus, hypothalamus, and
substantia nigra. The 1,25(OH)2D3 lowers oxidative stress, inhibits inflammation, offers
neuroprotection, down-regulates inflammatory mediators, and up-regulates neurotrophins
in neurons [62].
The Klotho gene, which was initially discovered as an ‘aging suppressor’ in mice,
encodes the antiaging protein Klotho. Klotho deficiency is linked to early mortality and
rapid aging, but its overexpression is linked to longevity. Klotho protein is involved in the
control of a number of biological processes, including calcium-phosphate balance, PTH,
and vit D metabolism [
63
]. The precise chemical pathways through which 1,25(OH)2D3 and
Klotho protein exerts its activities in the brain are yet unknown; however, the relationship
between them might be both genomic and non-genomic, but the interplay processes are
mainly unknown [63].
For all the above reasons, we could hypothesize that the therapeutic use of 1,25(OH)2D3
or similar agonists may have considerable promise due to its documented function in neu-
roinflammation, neurodegenerative diseases, and neuropsychiatric disorders.
Depression is a frequent mental illness in the elderly that lowers quality of life and
increases morbidity and death. Vit D may play a role in the onset and treatment of
depression as a neuro-steroid hormone [
64
]. One of the proposed mechanisms by which
this neurohormone exerts its action is its relationship with serotonin and dopamine levels
in the brain [65].
2.5. The Role of Vitamin D in Autoimmunity
Clinical studies have suggested that VDR polymorphisms and vit D deficiency is related
to the development and progression of several autoimmune diseases, such as rheumatoid
arthritis, systemic lupus erythematosus, multiple sclerosis, and autoimmune endocrine dis-
orders (e.g., Hashimoto thyroiditis, type-1 diabetes mellitus (T1DM), Addison’s disease,
and Graves’ disease) [
66
]. As mentioned, due to its great ability to bind to VDR and act
as a transcriptional factor, vit D may modulate gene expression and, consequently, exert
immunomodulatory effects on immune cells. Vit D has the capability to inhibit the produc-
tion of Th17 cytokine, improve Treg activity, stimulate NKT cell functions, inhibit Th1, and
induce the production of Th2 cytokine, and thereby shift T cells toward Th2 profile [
67
].
However, the role of vit D supplementation in the improvement of autoimmune diseases
remains unclear. Therefore, additional studies are needed to know the potential underlying
mechanisms involved [
68
]. Of note, all the clinical studies performed so far, only demonstrate
correlations. Thus, it is difficult to establish whether the low 25(OH)D3 level is the cause or
the consequence of autoimmune diseases [
69
]. Despite several studies having demonstrated a
beneficial effect of vit D supplementation in autoimmune diseases, there are also some studies
that did not show any effect on the main parameters of this kind of diseases. This could be
due to differences in the supplementation strategy or the individual characteristics of the
subjects included in the study, which are aspects that should be addressed in a properly way
at the moment of designing multicenter clinical trials [70,71].
3. Genetic and Epigenetic Regulation of Inflammation by Vitamin D
Epigenetics might explain the interplay between environment and genetics in the
development, progression, pathogenicity of disease, and the response to treatments. Epige-
netics changes occur in the following ways: DNA methylation, which generates methyl
cytosine in CpG dinucleotides sites leading to transcriptional silencing, histone modifica-
tions, chromatin remodeling, and noncoding RNAs regulation [
29
]. RNAs are untranslated
transcripts that play an important role in the post-transcriptional regulation of gene ex-
pression and can be classified into short, mid, and long based on their length. Among the
noncoding RNAs, long noncoding RNAs (lncRNAs) regulate gene expression by interacting
Nutrients 2023,15, 767 7 of 13
with DNA, messenger RNAs (mRNAs), and proteins, while microRNAs (miRNAs) mediate
the post-transcriptional repression or mRNA degradation in an epigenetic mechanism [
72
].
One of the most usual processes observed during vascular inflammation is histone
acetylation/deacetylation. Acetylation of histones (by histone acetyltransferases) is the
addition of positively charged acetyl groups to amino acid residues, which neutralizes the
negative charges of DNA reducing the affinity of histones to DNA, and leading to a more
relaxed chromatin state to allow higher accessibility to the transcriptional machinery [
73
].
Likewise, the opposite mechanism (deacetylation of histones) is carried out by histone
deacetylases (HDACs) [
74
]. Of interest, alterations in any of these systems may produce
abnormal activation or silencing of specific genes, which favors vascular disease onset. In
addition, transgenerational transmission of epigenetic signaling is informed to precede
vascular damage in children and young adults [
75
]. Thus, it is needed to investigate
the existing link between epigenetics and inflammation in the vasculature to understand
how epigenetic changes may influence these biological processes and how they could be
managed to inhibit the inflammatory cascade at the vascular level [76].
Notably, during pregnancy, most calcitriol is produced in the maternal kidneys and
a small proportion in the placenta [
77
]. Vit D induces epigenetic changes that modify the
way the placenta responds to this compound and its metabolites and can even reduce the
epigenetic changes associated with gestational aging and also plays a role in modifying
the immune response of the fetus [
78
]. Moreover, this research suggests that maternal
25(OH)D levels influence gene expression profiles, and these changes could contribute to
fetal immune imprinting and reducing allergic sensitization in the first years of life [78].
It is relevant to highlight that pregnant women are at an increased risk of developing vit
D deficiency, and this is associated with adverse health consequences among their offspring,
such as hypertension, hypocalcemia, deficient postnatal growth, and autoimmune diseases,
among others [
79
]. Regarding this, exposure of male and female Sprague-Dawley rats to
a vit D-free diet before mating led to an increase in systolic and diastolic blood pressure
in their offspring, in addition to the hypermethylation of the Pannexin-1 (Panx1), a gene
responsible for endothelial relaxation of large blood vessels [
80
]. Using the same model,
Zhang and coworkers also showed that maternal vitamin D-free diet (and ultraviolet-free
light) during pregnancy may cause insulin resistance in the offspring, which is accompanied
by persistent increased inflammation. The vit D-free diet caused a significant increase in
NFKBIA methylation at the CpG site +331 in the offspring. NFKBIA protein interacts with
REL dimers to block NF-kappa-B/REL complexes which are involved in inflammatory
pathways. Hence, through epigenetic modifications, the persistent lower I
κ
b
α
expression
levels in the progeny stimulates the activation of NF-
κ
B signaling which, in turn, leads
to inflammation with possible implication for the insulin resistance in adulthood [
81
].
These studies emphasize the relevance of vit D supplementation for the prevention of
epigenetically induced vascular inflammatory pathologies.
In fact, there is strong evidence that vit D and epigenetic mechanism programming is
highly related. As previously mentioned, calcitriol acts through its binding to the nuclear-
cytosolic VDR. After the heterodimer VDR-RXR is formed, genomic action is performed via
docking of the VDR-RXR complex to the VDRE in the proximal region of promoter genes
modulated by vitamin D [
82
,
83
]. The level of chromatin accessibility in the VDRE regions is
crucial for VDR target gene transcription. Hence, VDREs cannot be accessed by the active
VDR-RXR complex if it is inside a hypermethylated heterochromatin region. Therefore, the
transcriptional control of VDR-responsive genes is dependent on three mechanisms: vit D
availability, VDREs accessibility, and VDR expression levels. All these actions are regulated
by environmental, genetic, and epigenetic factors. From the environmental factors, sun
exposure, pollution, diet, and infection are able to regulate the VDR mainly by altering the
levels of vit D [84,85].
VDR expression is differently correlated with inflammatory cytokines expression
and miRNAs regulation, mainly miRNA-21, -214, and -125. A single nucleotide poly-
morphism of the VDR gene constitutes a risk factor for coronary artery disease [
86
]. In
Nutrients 2023,15, 767 8 of 13
macrophages, which are essential immune cells during the vascular inflammatory re-
sponse, upregulation of miRNA-155 is associated with LPS induced hyper-inflammatory
process by the inhibition of suppressor of cytokine signaling 1 (SOCS1). On the contrary,
1,25-dihydroxycholecalciferol-VDR signaling has been demonstrated to downregulate BIC
gene transcription by blocking the activation of NF-
κ
B, leading to decreased miRNA155 lev-
els and increased SOCS1 translation [
87
]. In addition, upregulation of epithelial VCAM-1,
ICAM-1, and IL-6 is related to inflammation and atherogenesis in an
in vivo
mouse model
of VDR deletion [
88
]. VDR is directly involved in the suppression of NF-
κ
B activation,
which may explain, at least in part, the VDR-mediated anti-inflammatory effects of vit D in
cardiovascular pathologies [
88
,
89
]. Most of the studies on vit D and vascular inflammatory
process focus on immune cells such as monocytes/macrophages or T cells activated by LPS.
In this context, Gynther and coworkers demonstrated that LPS-treated human monocytes
exposed to vit D (10 nM, cholecalciferol) downregulated the pro-inflammatory IL-12B gene
by VDR-RXR binding. By the recruitment of co-repressor NCOR2/SMRT and HDAC3,
a significant reduction in histone 4 acetylation and an increase in histone 3 trimethyla-
tion were observed at the IL-12B promoter [
90
]. Additionally, VDR-RXR binding to the
nuclear factor needed for the activation of T cell (NFAT) sites caused the dissociation
of acetylated histone H4 from IL17A promoter and recruitment of HDAC2 to the NFAT
sites, thus supporting the role of 1,25(OH)2D in attenuating the pathogenesis of vascular
inflammation [
91
]. By binding to the TLR4, LPS triggers mitogen-activated protein kinases
(MAPK) and NF-
κ
B signaling activation, causing the expression of TNF, IL1B, IL6, and
IL8 genes [
21
]. Curiously, when monocytes obtained from patients with type 1 diabetes
mellitus with microvascular complications were pre-incubated with vit D (0.1
µ
mol/L), a
reduction in their LPS-activated TLR4 expression and cytokine levels was observed [
92
].
Moreover, Zhang et al. [
93
] found that vit D dose dependently (15–70 ng/mL) inhibits
LPS-induced cytokine synthesis by the upregulation of MAPK phosphatase 1 (MKP-1) in
human monocytes and murine bone marrow-derived macrophages. Notably, increased
VDR binding to a putative VDRE in the MKP-1 promoter and histone H4 acetylation
close the VDRE site led to MAPK inactivation, thereby reducing p38 activation and cy-
tokine production. Another study demonstrated that 1,25(OH)2D has no distinguishable
effect on p38 phosphorylation in macrophages, suggesting that the downregulation of
the pro-inflammatory COX-2 happens in a MAPK-independent manner. In this study,
vit D blocked Akt/NF-
κ
B/COX-2 axis-mediated proinflammatory cytokines by binding
VDR to a functional VDRE in the thioesterase superfamily member 4 (THEM4) promoters,
thereby helping to the cardiovascular protective effects of vit D [
94
]. Epidemiological
studies informed that vit D deficiency (25(OH)D < 15 ng/ mL) could be considered a sign of
cardiovascular risk, mostly in people with hypertension [
95
]. In fact, there is an association
between vit D deficiency and inflammatory disorders in humans; this is so usual it is
considered a global issue [
96
]. Recently, Wimalawansa [
97
] recapitulated the role of vit
D on oxidative stress, epigenetics, gene expression, inflammation, and the aging process.
These aging-related processes seem to happen at lower rates in individuals with normal
vit D status. Severe vit D deficiency (25(OH)D
10 ng/mL) seems to be associated with
methylation in leukocyte DNA, mainly in MAPRE2 and DIO3 genes, both correlated with
tumor development [
98
]. All these studies support the genetic and epigenetic modulation
of vit D and its relevance in inflammation.
4. Conclusions
It is common to associate vit D with skeletal homeostasis. However, new roles are
constantly emerging for vit D with closer investigations. VDR is linking at hundreds of
sites in the genome and is associated with the regulation of more than 60 genes. Moreover,
vit D (and its active derivatives) not only play a role in the genetic regulation of many
genes, but also in the epigenetic regulation. In fact, the epigenetic machinery can be altered
by vit D, acting through multiple genetic mechanisms. Gene expression can be modified by
Nutrients 2023,15, 767 9 of 13
numerous miRNAs working as epigenetic modulators of VDR, also by methylation and
histone acetylation/deacetylation [2].
Vit D is involved in processes related to the development and progression of some
inflammatory diseases, and has the ability to alter serum levels of both proinflammatory
and anti-inflammatory mediators.
Beyond the known intimate relationship between vit D and the gastrointestinal tract,
we can highlight a bidirectional communication between the VDR and the intestinal micro-
biota. Both participate in an autoregulation in which different types of post-transcriptional
mechanisms and their relationship with intestinal permeability are also involved.
Moreover, vit D has protective effects in CKD and hypertension, and one of the mechanisms
by which it carries out this function is through the alteration of the RAAS system.
On the other hand, with new research findings, more insight is being obtained about
the relationship between vit D and different pathologies of the nervous system. Most
of these are focused on vit D and its relationship with oxidative stress. We know that
oxidative stress is one of the best-known starting points of different neuroinflammatory,
neurodegenerative, and neuropsychiatric processes. It is clear that vit D has implications
in general health and this fact invites researchers to continue looking for functions and
delving into the study of other physiological actions and how its deficiency is associated
with numerous pathologies beyond those explained here, such as diabetes, obesity, and
cancer. Thus, we speculate that vit D, now considered as calciotropic hormone, may later
be considered a hormone with multisystemic action.
Author Contributions:
Conceptualization, L.M. and W.M.; writing—original draft preparation, L.M.,
M.F.H., F.A.A. and W.M.; writing—review and editing, V.M.M.G. and S.G.M. All authors have read
and agreed to the published version of the manuscript.
Funding:
This research was funded by grants from Secretaría de Investigación, Internacionales y
Postgrado, Universidad Nacional de Cuyo (grant number 06/J044-T1), and from ANPCyT (Agencia
Nacional de Promoción de la Ciencia y la Tecnología, grant number PICT 2020 Serie A 4000), both
awarded to W. Manucha.
Institutional Review Board Statement: No application.
Informed Consent Statement: No application.
Data Availability Statement: No application.
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
Pop, T.L.; Sîrbe, C.; Ben¸ta, G.; Mititelu, A.; Grama, A. The role of vitamin D and vitamin D binding protein in chronic liver
Diseases. Int. J. Mol. Sci 2022,23, 10705. [CrossRef] [PubMed]
2.
Battistini, C.; Ballan, R.; Herkenhoff, M.E.; Saad, S.M.I.; Sun, J. Vitamin D modulates intestinal microbiota in inflammatory bowel
diseases. Int. J. Mol. Sci. 2020,22, 362. [CrossRef] [PubMed]
3. Holick, M.F. Vitamin D deficiency. N. Engl. J. Med. 2007,357, 266–281. [CrossRef] [PubMed]
4.
Slominski, A.T.; Chaiprasongsuk, A.; Janjetovic, Z.; Kim, T.K.; Stefan, J.; Slominski, R.M.; Hanumanthu, V.S.; Raman, C.; Qayyum,
S.; Song, Y.; et al. Photoprotective properties of vitamin D and lumisterol hydroxyderivatives. Cell Biochem. Biophys.
2020
,78,
165–180. [CrossRef]
5.
Giustina, A.; Bouillon, R.; Binkley, N.; Sempos, C.; Adler, R.A.; Bollerslev, J.; Dawson-Hughes, B.; Ebeling, P.R.; Feldman, D.;
Heijboer, A.; et al. Controversies in vitamin D: A statement from the third international conference. JBMR Plus
2020
,4, e10417.
[CrossRef]
6.
Holick, M.F.; Binkley, N.C.; Bischoff-Ferrari, H.A.; Gordon, C.M.; Hanley, D.A.; Heaney, R.P.; Murad, M.H.; Weaver, C.M.
Endocrine society. Evaluation, treatment, and prevention of vitamin D deficiency: An endocrine society clinical practice guideline.
J. Clin. Endocrinol. Metab. 2011,96, 1911–1930. [CrossRef]
7. Institute of Medicine. Dietary Reference Intakes for Calcium and Vitamin D; The National Academies Press: Washington, DC, USA,
2011.
8.
Bouillon, R.; Manousaki, D.; Rosen, C.; Trajanoska, K.; Rivadeneira, F.; Richards, J.B. The health effects of vitamin D supplementa-
tion: Evidence from human studies. Nat. Rev. Endocrinol. 2022,18, 96–110. [CrossRef]
Nutrients 2023,15, 767 10 of 13
9.
Charoenngam, N.; Holick, M.F. Immunologic effects of vitamin D on human health and disease. Nutrients
2020
,12, 2097.
[CrossRef]
10. Vojinovic, J. Vitamin D receptor agonists’ anti-inflammatory properties. Ann. N. Y. Acad. Sci. 2014,1317, 47–56. [CrossRef]
11.
Sirajudeen, S.; Shah, I.; Al Menhali, A. A narrative role of vitamin D and its receptor: With current evidence on the gastric tissues.
Int. J. Mol. Sci. 2019,20, 3832. [CrossRef]
12.
Charoenngam, N.; Shirvani, A.; Kalajian, T.A.; Song, A.; Holick, M.F. The effect of various doses of oral vitamin D
3
supplementa-
tion on gut microbiota in healthy adults: A randomized, double-blinded, dose-response study. Anticancer Res.
2020
,40, 551–556.
[CrossRef] [PubMed]
13.
Ferder, L.; Martín Giménez, V.M.; Inserra, F.; Tajer, C.; Antonietti, L.; Mariani, J.; Manucha, W. Vitamin D supplementation as a
rational pharmacological approach in the COVID-19 pandemic. Am. J. Physiol. Lung Cell. Mol. Physiol.
2020
,319, L941–L948.
[CrossRef] [PubMed]
14.
Martín Giménez, V.M.; Inserra, F.; Ferder, L.; García, J.; Manucha, W. Vitamin D deficiency in African Americans is associated
with a high risk of severe disease and mortality by SARS-CoV-2. J. Hum. Hypertens. 2021,35, 378–380. [CrossRef] [PubMed]
15.
Martín Giménez, V.M.; Lahore, H.; Ferder, L.; Holick, M.F.; Manucha, W. The little-explored therapeutic potential of nanoformula-
tions of 1,25-dihydroxyvitamin D
3
and its active analogs in prevalent inflammatory and oxidative disorders. Nanomedicine
2021
,
16, 2327–2330. [CrossRef] [PubMed]
16.
Ismailova, A.; White, J.H. Vitamin D, infections and immunity. Rev. Endocr. Metab. Disord.
2022
,23, 265–277. [CrossRef] [PubMed]
17.
White, J.H. Emerging roles of vitamin D-induced antimicrobial peptides in antiviral innate immunity. Nutrients
2022
,14, 284.
[CrossRef]
18.
Jain, S.K.; Micinski, D. Vitamin D upregulates glutamate cysteine ligase and glutathione reductase, and GSH formation, and
decreases ROS and MCP-1 and IL-8 secretion in high-glucose exposed U937 monocytes. Biochem. Biophys. Res. Commun.
2013
,
437, 7–11. [CrossRef] [PubMed]
19.
Câmara, A.B.; Brandão, I.A. The relationship between vitamin D deficiency and oxidative stress can be independent of age and
gender. Int. J. Vitam. Nutr. Res. 2021,91, 108–123. [CrossRef]
20.
Dzik, K.P.; Kaczor, J.J. Mechanisms of vitamin D on skeletal muscle function: Oxidative stress, energy metabolism and anabolic
state. Eur. J. Appl. Physiol. 2019,119, 825–839. [CrossRef]
21. Hii, C.S.; Ferrante, A. The non-genomic actions of vitamin D. Nutrients 2016,8, 135. [CrossRef] [PubMed]
22.
Max, D.; Brandsch, C.; Schumann, S.; Kühne, H.; Frommhagen, M.; Schutkowski, A.; Hirche, F.; Staege, M.S.; Stangl, G.I.
Maternal vitamin D deficiency causes smaller muscle fibers and altered transcript levels of genes involved in protein degradation,
myogenesis, and cytoskeleton organization in the newborn rat. Mol. Nutr. Food Res. 2014,58, 343–352. [CrossRef] [PubMed]
23. Gómez de Tejada Romero, M.J. Acciones extraóseas de la vitamina D. Rev. Osteoporos. Metab. Miner. 2014,6, 11–18. [CrossRef]
24.
de Las Heras, N.; Martín Giménez, V.M.; Ferder, L.; Manucha, W.; Lahera, V. Implications of oxidative stress and potential role of
mitochondrial dysfunction in COVID-19: Therapeutic effects of vitamin D. Antioxidants 2020,9, 897. [CrossRef]
25.
Martín Giménez, V.M.; Ferder, L.; Inserra, F.; García, J.; Manucha, W. Differences in RAAS/vitamin D linked to genetics and
socioeconomic factors could explain the higher mortality rate in African Americans with COVID-19. Ther. Adv. Cardiovasc. Dis.
2020,14, 1753944720977715. [CrossRef]
26.
Bellia, A.; Garcovich, C.; D’Adamo, M.; Lombardo, M.; Tesauro, M.; Donadel, G.; Gentileschi, P.; Lauro, D.; Federici, M.; Lauro, R.;
et al. Serum 25-hydroxyvitamin D levels are inversely associated with systemic inflammation in severe obese subjects. Intern.
Emerg. Med. 2013,8, 33–40. [CrossRef]
27.
Zakharova, I.; Klimov, L.; Kuryaninova, V.; Nikitina, I.; Malyavskaya, S.; Dolbnya, S.; Kasyanova, A.; Atanesyan, R.; Stoyan, M.;
Todieva, A.; et al. Vitamin D insufficiency in overweight and obese children and adolescents. Front. Endocrinol.
2019
,10, 103.
[CrossRef] [PubMed]
28.
Reyman, M.; Verrijn Stuart, A.A.; van Summeren, M.; Rakhshandehroo, M.; Nuboer, R.; De Boer, F.K.; Van Den Ham, H.J.;
Kalkhoven, E.; Prakken, B.; Schipper, H. Vitamin D deficiency in childhood obesity is associated with high levels of circulating
inflammatory mediators, and low insulin sensitivity. Int. J. Obes. 2014,38, 46–52. [CrossRef] [PubMed]
29.
Martín Giménez, V.M.; Chuffa, L.G.A.; Simão, V.A.; Reiter, R.J.; Manucha, W. Protective actions of vitamin D, anandamide and
melatonin during vascular inflammation: Epigenetic mechanisms involved. Life Sci. 2022,288, 120191. [CrossRef]
30.
Teixeira, T.M.; da Costa, D.C.; Resende, A.C.; Soulage, C.O.; Bezerra, F.F.; Daleprane, J.B. Activation of Nrf2-antioxidant signaling
by 1,25-dihydroxycholecalciferol prevents leptin-induced oxidative stress and inflammation in human endothelial cells. J. Nutr.
2017,147, 506–513. [CrossRef] [PubMed]
31.
Oma, I.; Andersen, J.K.; Lyberg, T.; Molberg, Ø.; Whist, J.; Fagerland, M.; Almdahl, S.; Hollan, I. Plasma vitamin D levels and
inflammation in the aortic wall of patients with coronary artery disease with and without inflammatory rheumatic disease. Scand.
J. Rheumatol. 2017,46, 198–205. [CrossRef]
32.
Tay, H.M.; Yeap, W.H.; Dalan, R.; Wong, S.C.; Hou, H.W. Increased monocyte-platelet aggregates and monocyte-endothelial
adhesion in healthy individuals with vitamin D deficiency. FASEB J. 2020,34, 11133–11142. [CrossRef] [PubMed]
33.
Poniedziałek-Czajkowska, E.; Mierzy ´nski, R. Could vitamin D be effective in prevention of preeclampsia? Nutrients
2021
,13,
3854. [CrossRef] [PubMed]
34.
Gouni-Berthold, I.; Berthold, H.K. Vitamin D and vascular disease. Curr. Vasc. Pharmacol.
2021
,19, 250–268. [CrossRef] [PubMed]
Nutrients 2023,15, 767 11 of 13
35.
Sorokin, V.; Vickneson, K.; Kofidis, T.; Woo, C.C.; Lin, X.Y.; Foo, R.; Shanahan, C.M. Role of vascular smooth muscle cell plasticity
and interactions in vessel wall inflammation. Front. Immunol. 2020,11, 599415. [CrossRef]
36.
Lee, A.S.; Jung, Y.J.; Thanh, T.N.; Lee, S.; Kim, W.; Kang, K.P.; Park, S.K. Paricalcitol attenuates lipopolysaccharide-induced
myocardial inflammation by regulating the NF-κB signaling pathway. Int. J. Mol. Med. 2016,37, 1023–1029. [CrossRef]
37.
Martínez-Moreno, J.M.; Herencia, C.; de Oca, A.M.; Díaz-Tocados, J.M.; Vergara, N.; Gómez-Luna, M.J.; López-Argüello, S.D.;
Camargo, A.; Peralbo-Santaella, E.; Rodríguez-Ortiz, M.E.; et al. High phosphate induces a pro-inflammatory response by
vascular smooth muscle cells and modulation by vitamin D derivatives. Clin. Sci. 2017,131, 1449–1463. [CrossRef]
38.
Diez, E.R.; Altamirano, L.B.; García, I.M.; Mazzei, L.; Prado, N.J.; Fornes, M.W.; Carrión, F.D.C.; Zumino, A.Z.P.; Ferder, L.;
Manucha, W. Heart remodeling and ischemia-reperfusion arrhythmias linked to myocardial vitamin d receptors deficiency in
obstructive nephropathy are reversed by paricalcitol. J. Cardiovasc. Pharmacol. Ther. 2015,20, 211–220. [CrossRef]
39.
Sanz, R.L.; Mazzei, L.; Manucha, W. Implications of the transcription factor WT1 linked to the pathologic cardiac remodeling
post-myocardial infarction. Clin. Investig. Arterioscler. 2019,31, 121–127. [CrossRef]
40.
Mizobuchi, M.; Morrissey, J.; Finch, J.L.; Martin, D.R.; Liapis, H.; Akizawa, T.; Slatopolsky, E. Combination therapy with an
angiotensin-converting enzyme inhibitor and a vitamin D analog suppresses the progression of renal insufficiency in uremic rats.
J. Am. Soc. Nephrol. JASN 2007,18, 1796–1806. [CrossRef]
41.
García, I.M.; Altamirano, L.; Mazzei, L.; Fornés, M.; Cuello-Carrión, F.D.; Ferder, L.; Manucha, W. Vitamin D receptor-modulated
Hsp70/AT1 expression may protect the kidneys of SHRs at the structural and functional levels. Cell Stress Chaperones
2014
,19,
479–491. [CrossRef]
42.
Sanz, R.; Mazzei, L.; Santino, N.; Ingrasia, M.; Manucha, W. Vitamin D-mitochondria cross-talk could modulate the signaling
pathway involved in hypertension development: A translational integrative overview. Clin. Investig. Arterioscler.
2020
,32,
144–155. [PubMed]
43.
Martorell, S.; Hueso, L.; Gonzalez-Navarro, H.; Collado, A.; Sanz, M.J.; Piqueras, L. Vitamin D receptor activation reduces
angiotensin-ii-induced dissecting abdominal aortic aneurysm in apolipoprotein E-knockout mice. Arterioscler. Thromb. Vasc. Biol.
2016,36, 1587–1597. [CrossRef] [PubMed]
44.
Wimalawansa, S.J. Vitamin D and cardiovascular diseases: Causality. J. Steroid Biochem. Mol. Biol.
2018
,175, 29–43. [CrossRef]
[PubMed]
45.
Bennett, A.L.; Lavie, C.J. Vitamin D metabolism and the implications for atherosclerosis. Adv. Exp. Med. Biol.
2017
,996, 185–192.
46.
Raed, A.; Bhagatwala, J.; Zhu, H.; Pollock, N.K.; Parikh, S.J.; Huang, Y.; Havens, R.; Kotak, I.; Guo, D.-H.; Dong, Y. Dose responses
of vitamin D3 supplementation on arterial stiffness in overweight African Americans with vitamin D deficiency: A placebo
controlled randomized trial. PLoS ONE 2017,12, e0188424. [CrossRef]
47. Sun, J.; Zhang, Y.G. Vitamin D receptor influences intestinal barriers in health and disease. Cells 2022,11, 1129. [CrossRef]
48.
Xiang, W.; Kong, J.; Chen, S.; Cao, L.-P.; Qiao, G.; Zheng, W.; Liu, W.; Li, X.; Gardner, D.G.; Li, Y.C. Cardiac hypertrophy in vitamin
D receptor knockout mice: Role of the systemic and cardiac renin-angiotensin systems. Am. J. Physiol. Endocrinol. Metab.
2005
,
288, E125–E132. [CrossRef]
49. Chau, Y.Y.; Kumar, J. Vitamin D in chronic kidney disease. Indian J. Pediatr. 2012,79, 1062–1068. [CrossRef]
50. Dornas, W.C.; Silva, M.E. Animal models for the study of arterial hypertension. J. Biosci. 2011,36, 731–737. [CrossRef]
51.
Mazzei, L.; García, M.; Calvo, J.P.; Casarotto, M.; Fornés, M.; Abud, M.A.; Cuello-Carrión, D.; Ferder, L.; Manucha, W. Changes in
renal WT-1 expression preceding hypertension development. BMC Nephrol. 2016,17, 34. [CrossRef]
52.
Mazzei, L.; Docherty, N.G.; Manucha, W. Mediators and mechanisms of heat shock protein 70 based cytoprotection in obstructive
nephropathy. Cell Stress Chaperones 2015,20, 893–906. [CrossRef] [PubMed]
53.
Gascon-Barré, M.; Huet, P.M. Apparent [3H]1,25-dihydroxyvitamin D3 uptake by canine and rodent brain. Am. J. Physiol.
1983
,
244, E266–E271. [CrossRef] [PubMed]
54.
Lee, P.W.; Selhorst, A.; Lampe, S.G.; Liu, Y.; Yang, Y.; Lovett-Racke, A.E. Neuron-specific vitamin d signaling attenuates microglia
activation and CNS autoimmunity. Front. Neurol. 2020,11, 19. [CrossRef] [PubMed]
55.
Menéndez, S.G.; Martín Giménez, V.M.; Holick, M.F.; Barrantes, F.J.; Manucha, W. COVID-19 and neurological sequelae: Vitamin
D as a possible neuroprotective and/or neuroreparative agent. Life Sci. 2022,297, 120464. [CrossRef]
56.
Bayat, M.; Kohlmeier, K.A.; Haghani, M.; Haghighi, A.B.; Khalili, A.; Bayat, G.; Hooshmandi, E.; Shabani, M. Co-treatment of
vitamin D supplementation with enriched environment improves synaptic plasticity and spatial learning and memory in aged
rats. Psychopharmacology 2021,238, 2297–2312. [CrossRef]
57.
Gáll, Z.; Székely, O. Role of vitamin D in cognitive dysfunction: New molecular concepts and discrepancies between animal and
human findings. Nutrients 2021,13, 3672. [CrossRef]
58.
Kim, H.A.; Perrelli, A.; Ragni, A.; Retta, F.; De Silva, T.M.; Sobey, C.G.; Retta, S.F. Vitamin D deficiency and the risk of
cerebrovascular disease. Antioxidants 2020,9, 327. [CrossRef]
59.
Cui, C.; Xu, P.; Li, G.; Qiao, Y.; Han, W.; Geng, C.; Liao, D.; Yang, M.; Chen, D.; Jiang, P. Vitamin D receptor activation regulates
microglia polarization and oxidative stress in spontaneously hypertensive rats and angiotensin II-exposed microglial cells: Role
of renin-angiotensin system. Redox Biol. 2019,26, 101295. [CrossRef]
60.
Guo, X.; Yuan, J.; Wang, J.; Cui, C.; Jiang, P. Calcitriol alleviates global cerebral ischemia-induced cognitive impairment by
reducing apoptosis regulated by VDR/ERK signaling pathway in rat hippocampus. Brain Res. 2019,1724, 146430. [CrossRef]
Nutrients 2023,15, 767 12 of 13
61.
Alessio, N.; Belardo, C.; Trotta, M.C.; Paino, S.; Boccella, S.; Gargano, F.; Pieretti, G.; Ricciardi, F.; Marabese, I.; Luongo, L.; et al.
Vitamin D deficiency induces chronic pain and microglial phenotypic changes in mice. Int. J. Mol. Sci.
2021
,22, 3604. [CrossRef]
62.
Lang, F.; Ma, K.; Leibrock, C.B. 1,25(OH)2D3 in brain function and neuropsychiatric disease. Neurosignals
2019
,27, 40–49.
[PubMed]
63.
Dërmaku-Sopjani, M.; Kurti, F.; Xuan, N.T.; Sopjani, M. Klotho-dependent role of 1,25(OH)2D3 in the brain. Neurosignals
2021
,29,
14–23. [PubMed]
64.
Zech, L.D.; Scherf-Clavel, M.; Daniels, C.; Schwab, M.; Deckert, J.; Unterecker, S.; Herr, A.S. Patients with higher vitamin D levels
show stronger improvement of self-reported depressive symptoms in psychogeriatric day-care setting. J. Neural. Transm.
2021
,
128, 1233–1238. [CrossRef]
65.
Seyedi, M.; Gholami, F.; Samadi, M.; Djalali, M.; Effatpanah, M.; Yekaninejad, M.S.; Hashemi, R.; Abdolahi, M.; Chamari,
M.; Honarvar, N.M. The effect of vitamin D3 supplementation on serum BDNF, dopamine, and serotonin in children with
attention-deficit/hyperactivity disorder. CNS Neurol. Disord. Drug Targets 2019,18, 496–501. [CrossRef] [PubMed]
66.
Fletcher, J.; Bishop, E.L.; Harrison, S.R.; Swift, A.; Cooper, S.C.; Dimeloe, S.K.; Raza, K.; Hewison, M. Autoimmune disease and
interconnections with vitamin D. Endocr. Connect. 2022,11, e210554. [CrossRef] [PubMed]
67.
Yang, C.Y.; Leung, P.S.; Adamopoulos, I.E.; Gershwin, M.E. The implication of vitamin D and autoimmunity: A comprehensive
review. Clin. Rev. Allergy Immunol. 2013,45, 217–226.
68.
Ao, T.; Kikuta, J.; Ishii, M. The effects of vitamin D on immune system and inflammatory diseases. Biomolecules
2021
,11, 1624.
[CrossRef]
69.
Bellan, M.; Andreoli, L.; Mele, C.; Sainaghi, P.P.; Rigamonti, C.; Piantoni, S.; De Benedittis, C.; Aimaretti, G.; Pirisi, M.; Marzullo, P.
Pathophysiological role and therapeutic implications of vitamin D in autoimmunity: Focus on chronic autoimmune diseases.
Nutrients 2020,12, 789. [CrossRef]
70.
Dankers, W.; Colin, E.M.; van Hamburg, J.P.; Lubberts, E. Vitamin D in autoimmunity: Molecular mechanisms and therapeutic
potential. Front. Immunol. 2017,7, 697. [CrossRef]
71.
Murdaca, G.; Tonacci, A.; Negrini, S.; Greco, M.; Borro, M.; Puppo, F.; Gangemi, S. Emerging role of vitamin D in autoimmune
diseases: An update on evidence and therapeutic implications. Autoimmun. Rev. 2019,18, 102350. [CrossRef]
72.
Statello, L.; Guo, C.J.; Chen, L.L.; Huarte, M. Gene regulation by long non-coding RNAs and its biological functions. Nat. Rev.
Mol. Cell Biol. 2021,22, 96–118. [PubMed]
73.
Barnes, C.E.; English, D.M.; Cowley, S.M. Acetylation & Co: An expanding repertoire of histone acylations regulates chromatin
and transcription. Essays Biochem. 2019,63, 97–107. [PubMed]
74.
Rafehi, H.; Balcerczyk, A.; Lunke, S.; Kaspi, A.; Ziemann, M.; Kn, H.; Okabe, J.; Khurana, I.; Ooi, J.; Khan, A.W.; et al. Vascular
histone deacetylation by pharmacological HDAC inhibition. Genome Res. 2014,24, 1271–1284. [CrossRef] [PubMed]
75.
Ambrosini, S.; Mohammed, S.A.; Lüscher, T.F.; Costantino, S.; Paneni, F. New mechanisms of vascular dysfunction in car-
diometabolic patients: Focus on epigenetics. High Blood Press. Cardiovasc. Prev. 2020,27, 363–371. [CrossRef] [PubMed]
76.
Zarzour, A.; Kim, H.W.; Weintraub, N.L. Epigenetic regulation of vascular diseases. Arterioscler. Thromb. Vasc. Biol.
2019
,39,
984–990. [CrossRef]
77. Wagner, C.L.; Hollis, B.W. The extraordinary metabolism of vitamin D. eLife 2022,11, e77539. [CrossRef]
78. Al-Garawi, A.; Carey, V.J.; Chhabra, D.; Mirzakhani, H.; Morrow, J.; Lasky-Su, J.; Qiu, W.; Laranjo, N.; Litonjua, A.A.; Weiss, S.T.
The role of vitamin D in the transcriptional program of human pregnancy. PLoS ONE 2016,11, e0163832. [CrossRef]
79.
Mulligan, M.L.; Felton, S.K.; Riek, A.E.; Bernal-Mizrachi, C. Implications of vitamin D deficiency in pregnancy and lactation. Am.
J. Obstet. Gynecol. 2010,202, e1–e9. [CrossRef]
80.
Meems, L.M.; Mahmud, H.; Buikema, H.; Tost, J.; Michel, S.; Takens, J.; Verkaik-Schakel, R.N.; Vreeswijk-Baudoin, I.; Mateo-Leach,
I.V.; van der Harst, P.; et al. Parental vitamin D deficiency during pregnancy is associated with increased blood pressure in
offspring via Panx1 hypermethylation. Am. J. Physiol. Heart Circ. Physiol. 2016,311, H1571. [CrossRef]
81.
Zhang, H.; Chu, X.; Huang, Y.; Li, G.; Wang, Y.; Li, Y.; Sun, C. Maternal vitamin D deficiency during pregnancy results in
insulin resistance in rat offspring, which is associated with inflammation and I
κ
b
α
methylation. Diabetologia
2014
,57, 2165–2172.
[CrossRef]
82.
Pike, J.W.; Meyer, M.B. The vitamin D receptor: New paradigms for the regulation of gene expression by 1,25-dihydroxyvitamin
D3. Rheum. Dis. Clin. N. Am. 2012,38, 13–27. [CrossRef]
83.
Izzo, M.; Carrizzo, A.; Izzo, C.; Cappello, E.; Cecere, D.; Ciccarelli, M.; Iannece, P.; Damato, A.; Vecchione, C.; Pompeo, F. Vitamin
D: Not just bone metabolism but a key player in cardiovascular diseases. Life 2021,11, 452. [CrossRef]
84.
Apprato, G.; Fiz, C.; Fusano, I.; Bergandi, L.; Silvagno, F. Natural epigenetic modulators of vitamin D receptor. Appl. Sci.
2020
,10,
4096. [CrossRef]
85.
Saccone, D.; Asani, F.; Bornman, L. Regulation of the vitamin D receptor gene by environment, genetics and epigenetics. Gene
2015,561, 171–180. [CrossRef] [PubMed]
86.
Ionova, Z.I.; Sergeeva, E.G.; Berkovich, O.A. Genetic and epigenetic factors regulating the expression and function of the vitamin
D receptor in patients with coronary artery disease. Russ. J. Cardiol. 2021,26, 4251. [CrossRef]
87.
Chen, Y.; Liu, W.; Sun, T.; Huang, Y.; Wang, Y.; Deb, D.K.; Yoon, D.; Kong, J.; Thadhani, R.; Li, Y.C. 1,25-Dihydroxyvitamin D
promotes negative feedback regulation of TLR signaling via targeting microRNA-155-SOCS1 in macrophages. J. Immunol. 2013,
190, 3687–3695. [CrossRef] [PubMed]
Nutrients 2023,15, 767 13 of 13
88.
Bozic, M.; Álvarez, Á.; de Pablo, C.; Sanchez-Niño, M.D.; Ortiz, A.; Dolcet, X.; Encinas, M.; Fernandez, E.; Valdivielso, J.M.
Impaired vitamin D signaling in endothelial cell leads to an enhanced leukocyte-endothelium interplay: Implications for
atherosclerosis development. PLoS ONE 2015,10, e0136863. [CrossRef]
89.
Al-Rasheed, N.M.; Al-Rasheed, N.M.; Bassiouni, Y.A.; Hasan, I.H.; Al-Amin, M.A.; Al-Ajmi, H.N.; Mohamad, R.A. Vitamin
D attenuates pro-inflammatory TNF-
α
cytokine expression by inhibiting NF-kB/p65 signaling in hypertrophied rat hearts. J.
Physiol. Biochem. 2015,71, 289–299. [CrossRef] [PubMed]
90.
Gynther, P.; Toropainen, S.; Matilainen, J.M.; Seuter, S.; Carlberg, C.; Väisänen, S. Mechanism of 1
α
,25-dihydroxyvitamin
D(3)-dependent repression of interleukin-12B. Biochim. Biophys. Acta 2011,1813, 810–818. [CrossRef]
91.
Joshi, S.; Pantalena, L.C.; Liu, X.K.; Gaffen, S.L.; Liu, H.; Rohowsky-Kochan, C.; Ichiyama, K.; Yoshimura, A.; Steinman, L.;
Christakos, S.; et al. 1,25-dihydroxyvitamin D(3) ameliorates Th17 autoimmunity via transcriptional modulation of interleukin-
17A. Mol. Cell. Biol. 2011,31, 3653–3669. [CrossRef]
92.
Devaraj, S.; Yun, J.M.; Duncan-Staley, C.R.; Jialal, I. Low vitamin D levels correlate with the proinflammatory state in type 1
diabetic subjects with and without microvascular complications. Am. J. Clin. Pathol. 2011,135, 429–433. [CrossRef]
93.
Zhang, Y.; Leung, D.Y.; Richers, B.N.; Liu, Y.; Remigio, L.K.; Riches, D.W.; Goleva, E. Vitamin D inhibits monocyte/macrophage
proinflammatory cytokine production by targeting MAPK phosphatase-1. J. Immunol. 2012,188, 2127–2135. [CrossRef]
94.
Wang, Q.; He, Y.; Shen, Y.; Zhang, Q.; Chen, D.; Zuo, C.; Qin, J.; Wang, H.; Wang, J.; Yu, Y. Vitamin D inhibits COX-2 expression
and inflammatory response by targeting thioesterase superfamily member 4. J. Biol. Chem.
2014
,289, 11681–11694. [CrossRef]
[PubMed]
95.
Wang, T.J.; Pencina, M.J.; Booth, S.L.; Jacques, P.F.; Ingelsson, E.; Lanier, K.; Benjamin, E.J.; D’Agostino, R.B.; Wolf, M.; Vasan, R.S.
Vitamin D deficiency and risk of cardiovascular disease. Circulation 2008,117, 503–511. [CrossRef] [PubMed]
96. Norman, P.E.; Powell, J.T. Vitamin D and cardiovascular disease. Circ. Res. 2014,114, 379–393. [CrossRef]
97.
Wimalawansa, S.J. Vitamin D deficiency: Effects on oxidative stress, epigenetics, gene regulation, and aging. Biology
2019
,8, 30.
[CrossRef] [PubMed]
98. Zhu, H.; Wang, X.; Shi, H.; Su, S.; Harshfield, G.A.; Gutin, B.; Snieder, H.; Dong, Y. A genome-wide methylation study of severe
vitamin D deficiency in African American adolescents. J. Pediatr. 2013,162, 1004–1009.e1. [CrossRef] [PubMed]
Disclaimer/Publisher’s Note:
The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.
... The VDR, belonging to the nuclear receptor superfamily of transcription factors, mediates genomic effects by regulating gene expression linked to cell survival and anti-inflammatory response. However, the precise role of the VDR in brain pathologies remains an active area of research due to the multi-target mechanisms of vitamin D and the overlap of its genomic and non-genomic effects [34,35]. Moreover, preclinical studies have shown that vitamin D treatments can reverse behavioral changes, reduce oxidative stress, and decrease pro-inflammatory markers, strengthening the hypothesis that vitamin D offers neuroprotection and could be beneficial in preventing or treating neurodegenerative diseases [34]. ...
... As mentioned before, upon binding to a vitamin D response element (DRE), the VDR orchestrates the expression of multiple genes implicated in inflammation [17]. Vitamin D contributes to a reduction in the production of pro-inflammatory cytokines such as TNF-α, interleukin 1β (IL-1β), and interleukin 6 (IL-6), which are implicated in the pathogenesis of diseases such as Alzheimer's, Parkinson's, and multiple sclerosis [35,39]. Concurrently, it enhances the secretion of interleukin 10 (IL-10), an anti-inflammatory cytokine that safeguards brain tissue from inflammatory damage [40]. ...
... Furthermore, vitamin D's role extends to influencing Transforming Growth Factor beta 1 (TGF-β1), which is involved in the regulation of inflammation and fibrosis [35], and modulating Matrix Metalloproteinases (MMPs), enzymes critical in extracellular matrix remodeling [43]. The VDR's involvement in gene regulation also extends to programmed cell death, highlighting the diverse mechanisms by which vitamin D can contribute to neuroprotection and potentially the prevention and treatment of neurodegenerative diseases [44]. ...
Article
Full-text available
Neuroinflammation represents a critical immune response within the brain, playing a pivotal role in defense against injury and infection. However, when this response becomes chronic, it can contribute to the development of various neurodegenerative and psychiatric disorders. This bibliographic review delves into the role of vitamin D in modulating neuroinflammation and its implications for brain health, particularly in the context of neurological and psychiatric disorders. While vitamin D is traditionally associated with calcium homeostasis and bone health, it also exerts immunomodulatory and neuroprotective effects within the central nervous system. Through comprehensive analysis of preclinical and clinical studies, we uncover how vitamin D, acting through its receptors in glial cells, may influence the production of proinflammatory cytokines and antioxidants, potentially mitigating the cascade of events leading to neuronal damage. Clinical research has identified vitamin D deficiency as a common thread in the increased risks of multiple sclerosis, Parkinson's disease, Alzheimer's, and depression, among others. Furthermore, preclinical models suggest vitamin D's regulatory capacity over inflammatory mediators, its protective role against neuronal apoptosis, and its contribution to neurogenesis and synaptic plasticity. These insights underscore the potential of vitamin D supplementation not only in slowing the progression of neurodegenerative diseases but also in improving the quality of life for patients suffering from psychiatric conditions. Future clinical studies are essential to validate these findings and further our understanding of vitamin D's capacity to prevent or alleviate symptoms, opening new avenues for therapeutic strategies against neuroinflammation-related pathologies.
... Furthermore, few studies attempt to investigate treatments for long post-COVID-19 syndrome for which there is no evidence of efficacy and little biological plausibility. Extra-Skeletal functions of VD including differentiation and proliferation of cells, antioxidant, antibacterial, anti-inflammatory and immunomodulatory properties have been investigated in various tissues or cells by many investigators (Holick et al. 2023). Several epidemiologic studies have observed that low VD levels are found in a large percentage of COVID-19 patients with acute respiratory failure. ...
... Therefore, patients with severe COVID-19 may benefit from vitamin D supplementation, which would help reduce the production of IL-6 that causes a cytokine storm and thus reduce the severity of the disease. (Azmi et al. 2023;Chileshe et al. 2022;Holick et al. 2023). Furthermore, calcitriol has been shown to improve the barrier function of 16HBE cell layers based on two independent measures by inhibiting TNF-α-induced barrier leakage of epithelial cells in a human lung culture model (Rybakovsky et al. 2023). ...
Article
Full-text available
Nearly four years after its first appearance, and having gone from pandemic to endemic, the SARS-CoV-2 remains out of control globally. The purpose of this study was to evaluate the clinical efficacy of vitamin D (VD) in COVID-19 and long COVID-19, explain the discrepancy in clinical outcomes and highlight the potential impact of metformin on VD efficacy in recent articles. Articles from January 2022 to August 2023 were selected for this review. The objective of this study was achieved by reviewing, analyzing, and discussing articles demonstrating (1) the mechanism of action of VD (2) observational or randomized clinical trials (RCTs) that support or not the beneficial clinical effects of VD in COVID-19 or long COVID. (3) genetic and non-genetic reasons for the variation in the effects of VD. Articles were collected from electronic databases such as PubMed, Scopus, MEDLINE, Google Scholar, Egyptian Knowledge Bank, Science Direct, and Cochrane Database of Systematic Reviews. Twenty three studies conducted in vitro or in animal models indicated that VD may act in COVID-19 through protecting the respiratory system by antimicrobial peptide cathelicidins, reducing lung inflammation, regulating innate and adaptive immune functions and up regulation of autophagy gene activity. Our review identified 58 clinical studies that met the criteria. The number of publications supporting a beneficial clinical activity of VD in treating COVID-19 was 49 (86%), including 12 meta-analyses. Although the total patients included in all articles was 14,071,273, patients included in publications supporting a beneficial role of VD in COVID-19 were 14,029,411 (99.7%). Collectively, extensive observational studies indicated a decisive relationship between low VD levels and the severity of COVID-19 and mortality outcomes. Importantly, evidence from intervention studies has demonstrated the effectiveness of VD supplements in treating COVID-19. Furthermore, the results of 4 observational studies supported the beneficial role of VD in alleviating symptoms of long COVID-19 disease. However, eight RCTs and one meta-analysis of RCTs may contain low-grade evidence against a beneficial role of VD in COVID-19. Twenty-five articles have addressed the association between VDR and DBP genetic polymorphisms and treatment failure of VD in COVID-19. Impaired VDR signaling may underlie the variability of VD effects as non-genetic mechanisms. Interestingly, in recent studies, metformin has a beneficial therapeutic role in COVID-19 and long COVID-19, possibly by improving AMPK signaling of the VDR and enhancing the efficacy of the VD. In conclusion, evidence has been significantly strengthened over the past 18 months, with several meta-analyses and RCTs reporting conclusive beneficial effects of VD supplementation against COVID-19 and highlighting metformin to improve VDR sensitivity and efficacy in treating COVID-19 and long COVID-19.
... Similar mechanisms regulating the intracrine, extra-renal conversion of 25(OH)D to 1,25(OH) 2 D have been suggested in many other tissues and cells, such as cancer cells, placenta, mammary glands, immune cells, and osteoblasts [9]. The autocrine and paracrine action resulting from the locally activated vitamin D, together with the expression of its receptor (vitamin D receptor, VDR) in many tissues, has been linked to its pleiotropic effects, particularly those on the immune and cardiovascular system, cell proliferation, and muscle function [14,15]. ...
... As universally accepted, vitamin D deficiency is a common condition in the general population, and in particular in subjects with chronic disorders [15]. The knowledge acquired from many studies, as well as clinical practice, on the high prevalence of hypovitaminosis D in many metabolic chronic diseases, including those related to mineral homeostasis, allows us to speculate that it could be an issue in HypoPT, as well. ...
Article
Full-text available
Hypoparathyroidism (HypoPT) is a rare endocrine disorder characterized by the absence or insufficient parathyroid hormone production resulting in chronic hypocalcemia. Complications of HypoPT include perturbation of several target organs. The conventional treatment consists of the administration of active vitamin D, namely calcitriol. Regarding vitamin D status, few data are available, mostly in HypoPT subjects supplemented with parent vitamin D. In addition, perturbation of vitamin D metabolism has been poorly investigated, as well as the contribution of altered vitamin D status on the clinical expression of the disease. The most recent consensus on the management of chronic HypoPT suggests the baseline evaluation of serum 25-hydroxy-vitamin D [25(OH)D] and supplementation with parent vitamin D with the aim to achieve and maintain serum 25(OH)D levels in the range of 30–50 ng/mL. The rationale for using supplementation with parent vitamin D (either ergocalciferol or cholecalciferol) in HypoPT would be to provide sufficient 25(OH)D substrate to the residual 1-α-hydroxylase activity, thus ensuring its conversion to active vitamin D in renal and extra-renal tissues. More data from experimental and clinical studies are needed for better assessing how these mechanisms may significantly influence metabolic control in HypoPT and eventually skeletal and extra-skeletal manifestation of the disease. Finally, future data will clarify how the currently available parent vitamin D compounds (ergocalciferol, cholecalciferol, calcifediol) would perform in addressing these specific issues.
... In the 12 studies included, we observed that lower baseline 25OHD levels increased the risk of developing diabetes by Figure 4 shows the association between lower baseline serum 25OHD levels and incident diabetes at follow-up after adjusting for potential confounders. The median number of adjustments in the multivariable analyses was 11 (range: [4][5][6][7][8][9][10][11][12][13][14][15][16][17], with all the studies including adiposity estimates, as fully detailed in Table 1. In the 12 studies included, we observed that lower baseline 25OHD levels increased the risk of developing diabetes by 19% (HR = 1.22; 95% CI: 1.12-1.33; ...
Article
Full-text available
Vitamin D deficiency is very common worldwide, particularly in old age, when people are at the highest risk of the negative adverse consequences of hypovitaminosis D. Additionally to the recognized functions in the regulation of calcium absorption, bone remodeling, and bone growth, vitamin D plays a key role as a hormone, which is supported by various enzymatic, physiological, metabolic, and pathophysiological processes related to various human organs and systems. Accruing evidence supports that vitamin D plays a key role in pancreatic islet dysfunction and insulin resistance in type 2 diabetes. From an epidemiological viewpoint, numerous studies suggest that the growing incidence of type 2 diabetes in humans may be linked to the global trend of prevalent vitamin D insufficiency. In the past, this association has raised discussions due to the equivocal results, which lately have been more convincing of the true role of vitamin D supplementation in the prevention of incident type 2 diabetes. Most meta-analyses evaluating this role have been conducted in adults or young older persons (50–60 years old), with only one focusing on older populations, even if this is the population at greater risk of both hypovitaminosis D and type 2 diabetes. Therefore, we conducted an update of the previous systematic review and meta-analysis examining whether hypovitaminosis D (low serum 25OHD levels) can predict incident diabetes in prospective longitudinal studies among older adults. We found that low 25OHD was associated with incident diabetes in older adults even after adjusting for several relevant potential confounders, confirming and updating the results of the only previous meta-analysis conducted in 2017.
... In conclusion, the beneficial effects on the nervous system cells are enhanced because a significant amount of substance can reach both neurons and Schwann cells due to the combined cooperative effect of NAC, VitD3, and Glut. Indeed, NAC protects cells from oxidative stress and improves glutathione availability [63], and the presence of VitD3 supports the optimal mitochondrial function reduction in the inflammatory network due to its ability to induce genomic and non-genomic effects [18,64]. ...
Article
Full-text available
Chronic oxidative stress has been consistently linked to age-related diseases, conditions, and degenerative syndromes. Specifically, the brain is the organ that significantly contributes to declining quality of life in ageing. Since the body cannot completely counteract the detrimental effects of oxidative stress, nutraceuticals’ antioxidant properties have received significant attention in recent years. This study assesses the potential health benefits of a novel combination of glutathione, vitamin D3, and N-acetylcysteine. To examine the combination’s absorption and biodistribution and confirm that it has no harmful effects, the bioavailability of the mixture was first evaluated in a 3D model that mimicked the intestinal barrier. Further analyses on the blood–brain barrier was conducted to determine the antioxidant effects of the combination in the nervous system. The results show that the combination reaches the target and successfully crosses the blood–brain and intestinal barriers, demonstrating enhanced advantages on the neurological system, such as a reduction (about 10.5%) in inflammation and enhancement in cell myelination (about 20.4%) and brain tropism (about 18.1%) compared to the control. The results support the cooperative effect of N-acetylcysteine, vitamin D3, and glutathione to achieve multiple health benefits, outlining the possibility of an alternative nutraceutical approach.
... As the common active form of vitamin D, 1,25(OH) 2 D 3 impacts multiple biological processes, such as cell differentiation and immune and inflammatory responses [14]. Besides its crucial anti-inflammatory function, vitamin D has beneficial effects on suppressing reactive oxygen species (ROS) and nitric oxide, which may restrain oxidative damage [15]. Vitamin D could regulate insulin, restore pancreatic β-cell function, and suppress cell apoptosis and oxidative stress in T2DM rat models [14]. ...
Article
Full-text available
Objective Type 2 diabetes mellitus (T2DM) is a common metabolic disorder with rising incidence worldwide. This study explored the anti-T2DM role of vitamin D, thereby providing novel therapeutic strategies. Methods C57BL/6 J mice and MIN6 cells were used to induce in vivo T2DM and damaged β-cell models, respectively. Body weights, fasting blood glucose, and fasting insulin were measured in mice. Oral glucose tolerance test (OGTT) and insulin tolerance test (ITT) were conducted on mice. Lipid indices (TG, TC, LDL-C, and HDL-C) were detected in mouse serum. Hematoxylin-eosin staining was used to evaluate pancreatic tissue injury. ELISA was used to assess insulin and oxidative stress (OS) markers (MDA, GSH, and SOD) in mice and MIN6 cells. Production of ROS was detected in islet β-cells and MIN6 cells. Cell viability and apoptosis were evaluated using CCK-8 and flow cytometry, respectively. QRT-PCR and western blotting were used to detect pro-inflammatory factors (TNF-α and IL-6) and endoplasmic reticulum stress (ERS) markers (CHOP and GRP78), respectively. Results Vitamin D reduced body weights, fasting blood glucose, and insulin and ameliorated glucose tolerance and insulin sensitivity in T2DM mice. Besides, vitamin D decreased serum TG, TC, LDL-C, and increased HDL-C in T2DM mice. Vitamin D inhibited pancreatic histopathological injury, cell apoptosis, OS, and β-cell decline in T2DM mice. Moreover, vitamin D alleviated cell death, insufficient insulin secretion, inflammation, OS, and ERS in damaged MIN6 cells. Notably, N-acetyl-L-cysteine (an OS inhibitor) enhanced these effects of vitamin D. Conclusions Vitamin D relieved T2DM symptoms by alleviating OS-induced β-cell impairment.
... VitD could support the immune defense at s25(OH)D levels above 50 ng/mL in the fight against infection [severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection] and thus prevent potentially stressful situations [201][202][203] and should be incorporated into the medical treatment plan. In "lifelong" autoimmune processes in progressive MS, the immunomodulatory activity of vitD can only reduce excessive inflammatory damage if optimal levels of s25(OH)D are sustainably achieved. ...
Article
Full-text available
Over the past decade, knowledge of the pathophysiology and immunology of multiple sclerosis (MS) and depression, and the complex links to vitamin D (VitD) balance, has increased rapidly. Both diseases are characterized by an imbalance of proinflammatory and antiinflammatory cytokines, increased serum neurofilament light chains (sNfLs), disruption of the blood-brain barrier (BBB), abolition of the physiological function of the various types of microglia (MG), decreased calcidiol-serum levels, and disorders of the gut microbiome in combination with hyperactivity of the hypothalamic-pituitary-adrenal (HPA)-axis/microbiome-gut-brain-axis characterized. In depression, stress initiates cellular and molecular changes in the brain via increased cortisol release in the HPA-axis. Microglial activation and neuronal damage as well as dysregulation of neuroplastic and neurotrophic factors complete the spectrum of pathological damage. It is shown that gut dysbiosis leads to increased gut permeability, which favors endotoxemia and ultimately paves the way to systemic inflammation. A VitD supplementation could restore the balance of microorganisms in the intestine and reduce the inflammatory processes at various levels. VitD promotes regulatory T cell (Treg) proliferation, inhibits the expression of T helper 1 (Th1) cells and Th17 immune cells, and inhibits proinflammatory interleukin-17 (IL-17). 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] reduces also the secretion of interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α). Increased calcitriol levels lead to a reduction in MG activation, oxidative stress, and lower BBB permeability. An early, permanent, daily sufficient VitD supplementation as an add-on therapy under control of the serum 25-hydroxyvitamin D [s25(OH)D] levels is an essential therapeutic tool to slow down the disability caused by MS and thereby primarily prevent or reduce the stress and subsequently the manifestation of depression. Through the future continuous measurement of the biomarkers serum neurofilament ligth chains and glial fibrillary acidic proteins as well as the s25(OH)D level in MS and comorbidity depression, future therapy successes or failures can be avoided.
... In this context, vitamin D and some of its analogs (e.g., paricalcitol) can significantly reduce serum levels of proinflammatory vascular markers such as interleukins 1, 2, 6 and 1β (IL-1, IL-2, IL-6 and IL-1β), tumor necrosis factor-alpha (TNF-α), high-sensitivity C reactive protein (hCRP), fibrinogen, C-C Motif Chemokine Ligand 2 and 5 (CCL2 and CCL5), transforming growth factor beta (TGF-β), fractalkine (a chemoattractant cytokine), prostaglandin E2 (PG-E2), vascular endothelial growth factor (VEGF), C-X-C Motif Chemokine Ligand 1 (CXCL1), and interferon-gamma ((IFN-ϒ), which is responsible for the activation of T-helper-1 (Th1) cells), among others ( Fig. 1) [2]. Vitamin D is also able to decrease endothelial tissue expression of vascular cell adhesion molecule 1 (VCAM-1), intercellular adhesion mol-In addition, it is essential to highpoint that both endothelial and vascular smooth muscle cells (VSMCs) highly express vitamin D receptors (VDR), reinforcing vitamin D's crucial role in attenuating vascular inflammation. ...
Article
Full-text available
Drug discovery and development are usually very costly and time-consuming. Therefore, drug repurposing is an attractive option for treating multiple diseases due to significantly reducing the time and costs invested in these tasks while using less risky compounds. Moreover, drug repurposing may help to discover new drug targets and signaling pathways that may be therapeutically exploited. Such is the case of vitamin D (or hormone D), which has recently gained relevance in clinical and experimental medicine with an impact on pathologies other than the phosphocalcic mechanism. In this sense, there is accumulated evidence that supports that -from now on- vitamin D (or more recently called hormone D) should be recognized both for its vascular anti-inflammatory actions as well as its bone effects. Likewise, this compound could be proposed as a new therapeutic alternative in the prevention and treatment of hypertension, atherosclerosis, and many other vascular inflammatory diseases due to the possibility of being used as a repurposed drug.
Article
Full-text available
Vitamin D (calciferol) is a fat-soluble vitamin that has a significant role in phospho-calcium metabolism, maintaining normal calcium levels and bone health development. The most important compounds of vitamin D are cholecalciferol (vitamin D3, or VD3) and ergocalciferol (vitamin D2, or VD2). Besides its major role in maintaining an adequate level of calcium and phosphate concentrations, vitamin D is involved in cell growth and differentiation and immune function. Recently, the association between vitamin D deficiency and the progression of fibrosis in chronic liver disease (CLD) was confirmed, given the hepatic activation process and high prevalence of vitamin D deficiency in these diseases. There are reports of vitamin D deficiency in CLD regardless of the etiology (chronic viral hepatitis, alcoholic cirrhosis, non-alcoholic fatty liver disease, primary biliary cirrhosis, or autoimmune hepatitis). Vitamin D binding protein (VDBP) is synthesized by the liver and has the role of binding and transporting vitamin D and its metabolites to the target organs. VDBP also plays an important role in inflammatory response secondary to tissue damage, being involved in the degradation of actin. As intense research during the last decades revealed the possible role of vitamin D in liver diseases, a deeper understanding of the vitamin D, vitamin D receptors (VDRs), and VDBP involvement in liver inflammation and fibrogenesis could represent the basis for the development of new strategies for diagnosis, prognosis, and treatment of liver diseases. This narrative review presents an overview of the evidence of the role of vitamin D and VDBP in CLD, both at the experimental and clinical levels.
Article
Full-text available
Vitamin D receptor (VDR) executes most of the biological functions of vitamin D. Beyond this, VDR is a transcriptional factor regulating the expression levels of many target genes, such as genes for tight junction proteins claudin-2, -5, -12, and -15. In this review, we discuss the progress of research on VDR that influences intestinal barriers in health and disease. We searched PubMed and Google Scholar using key words vitamin D, VDR, tight junctions, cancer, inflammation, and infection. We summarize the literature and progress reports on VDR regulation of tight junction distribution, cellular functions, and mechanisms (directly or indirectly). We review the impacts of VDR on barriers in various diseases, e.g., colon cancer, infection, inflammatory bowel disease, and chronic inflammatory lung diseases. We also discuss the limits of current studies and future directions. Deeper understanding of the mechanisms by which the VDR signaling regulates intestinal barrier functions allow us to develop efficient and effective therapeutic strategies based on levels of tight junction proteins and vitamin D/VDR statuses for human diseases.
Article
Full-text available
Vitamin D helps the intestine to absorb calcium and other minerals that the body needs, and provides support to the immune system. To carry out these roles, vitamin D must be converted into the active hormone calcitriol (also known as 1,25-dihydroxy-vitamin D). First, vitamin D is metabolized by the liver into a compound called 25(OH)D, which then is broken down into its active form calcitriol, mainly in the kidneys. This metabolic process is tightly regulated and relies on calcium and various hormones, including calcitriol itself (Pike and Christakos, 2017).
Article
Full-text available
Vitamin D has well documented effects on calcium homeostasis and bone metabolism but recent studies suggest a much broader role for this secosteroid in human health. Key components of the vitamin D system, notably the vitamin D receptor (VDR) and the vitamin D activating enzyme (1α-hydroxylase), are present in a wide array of tissues, notably macrophages, dendritic cells and T lymphocytes (T cells) from the immune system. Thus, serum 25-hydroxyvitamin D (25D) can be converted to hormonal 1,25-dihydroxyvitamin D (1,25D) within immune cells, and then interact with VDR and promote transcriptional and epigenomic responses in the same or neighbouring cells. These intracrine and paracrine effects of 1,25D have been shown to drive antibacterial or antiviral innate responses, as well as attenuating inflammatory T cell adaptive immunity. Beyond these mechanistic observations, association studies have reported correlation between low serum 25D levels and the risk and severity of human immune disorders including autoimmune diseases such as inflammatory bowel disease, multiple sclerosis, type 1 diabetes and rheumatoid arthritis. The proposed explanation for this is that decreased availability of 25D compromises immune cell synthesis of 1,25D leading to impaired innate immunity and over-exuberant inflammatory adaptive immunity. The aim of the current review is to explore the mechanistic basis for immunomodulatory effects of 25D and 1,25D in greater detail with specific emphasis on how vitamin D-deficiency (low serum levels of 25D) may lead to dysregulation of macrophage, dendritic cell and T cell function, and increase risk of inflammatory autoimmune disease.
Article
Full-text available
Vitamin D deficiency, characterized by low circulating levels of calcifediol (25-hydroxyvitamin D, 25D) has been linked to increased risk of infections of bacterial and viral origin. Innate immune cells produce hormonal calcitriol (1,25-dihydroxyvitamin D, 1,25D) locally from circulating calcifediol in response to pathogen threat and an immune-specific cytokine network. Calcitriol regulates gene expression through its binding to the vitamin D receptor (VDR), a ligand-regulated transcription factor. The hormone-bound VDR induces the transcription of genes integral to innate immunity including pattern recognition receptors, cytokines, and most importantly antimicrobial peptides (AMPs). Transcription of the human AMP genes β-defensin 2/defensin-β4 (HBD2/DEFB4) and cathelicidin antimicrobial peptide (CAMP) is stimulated by the VDR bound to promoter-proximal vitamin D response elements. HDB2/DEFB4 and the active form of CAMP, the peptide LL-37, which form amphipathic secondary structures, were initially characterized for their antibacterial actively. Notably, calcitriol signaling induces secretion of antibacterial activity in vitro and in vivo, and low circulating levels of calcifediol are associated with diverse indications characterized by impaired antibacterial immunity such as dental caries and urinary tract infections. However, recent work has also provided evidence that the same AMPs are components of 1,25D-induced antiviral responses, including those against the etiological agent of the COVID-19 pandemic, the SARS-CoV2 coronavirus. This review surveys the evidence for 1,25D-induced antimicrobial activity in vitro and in vivo in humans and presents our current understanding of the potential mechanisms by which CAMP and HBD2/DEFB4 contribute to antiviral immunity.
Article
Coronary artery disease (CAD) remains the leading cause of death and disability in developed countries. Using traditional risk factors for CAD, it is possible to predict the likelihood of acute coronary events in no more than 50% of cases. Therefore, the study of influence of genetic and epigenetic factors on the development of CAD is extremely important. Research in recent years has shown that vitamin D deficiency is a new risk factor for atherosclerosis and immune inflammation. Vitamin D implements protective effects against immune inflammation through receptors in the vascular wall. A single nucleotide polymorphism of the vitamin D receptor (VDR) gene is a potential risk factor for CAD associated with low vitamin D levels. VDR expression correlates with the expression of pro-inflammatory cytokines and is regulated by microRNAs — microRNA-125a-5p, microRNA-125b-5p, microRNA-214-3p and microRNA-21 These microRNAs regulate the action, synthesis and metabolism of vitamin D and can themselves be influenced by VDR signals through dynamic feedback, which can lead to destabilization of mRNA and inhibition of translation. This literature review highlights the effect of a single nucleotide polymorphism of the VDR gene and microRNA on the pathogenetic mechanisms of CAD.
Article
SARS-CoV-2, the etiological agent of the current COVID-19 pandemic, belongs to a broad family of coronaviruses that also affect humans. SARS-CoV-2 infection usually leads to bilateral atypical pneumonia with significant impairment of respiratory function. However, the infectious capacity of SARS-CoV-2 is not limited to the respiratory system, but may also affect other vital organs such as the brain. The central nervous system is vulnerable to cell damage via direct invasion or indirect virus-related effects leading to a neuroinflammatory response, processes possibly associated with a decrease in the activity of angiotensin II converting enzyme (ACE2), the canonical cell-surface receptor for SARS-CoV-2. This enzyme regulates neuroprotective and neuroimmunomodulatory functions and can neutralize both inflammation and oxidative stress generated at the cellular level. Furthermore, there is evidence of an association between vitamin D deficiency and predisposition to the development of severe forms of COVID-19, with its possible neurological and neuropsychiatric sequelae: vitamin D has the ability to down-modulate the effects of neuroinflammatory cytokines, among other anti-inflammatory/immunomodulatory effects, thus attenuating harmful consequences of COVID-19. This review critically analyzes current evidence supporting the notion that vitamin D may act as a neuroprotective and neuroreparative agent against the neurological sequelae of COVID-19.
Article
Vascular inflammation is one of the main activating stimuli of cardiovascular disease and its uncontrolled development may worsen the progression and prognosis of these pathologies. Therefore, the search for new therapeutic options to treat this condition is undoubtedly needed. In this regard, it may be better to repurpose endogenous anti-inflammatory compounds already known, in addition to synthesizing new compounds for therapeutic purposes. It is well known that vitamin D, anandamide, and melatonin are promising endogenous substances with powerful and widespread anti-inflammatory properties. Currently, the epigenetic mechanisms underlying these effects are often unknown. This review summarizes the potential epigenetic mechanisms by which vitamin D, anandamide, and melatonin attenuate vascular inflammation. This information could contribute to the improvement in the therapeutic management of multiple pathologies associated with blood vessel inflammation, through the pharmacological manipulation of new target sites that until now have not been addressed.
Article
Vitamin D supplementation can prevent and cure nutritional rickets in infants and children. Preclinical and observational data suggest that the vitamin D endocrine system has a wide spectrum of skeletal and extra-skeletal activities. There is consensus that severe vitamin D deficiency (serum 25-hydroxyvitamin D (25OHD) concentration <30 nmol/l) should be corrected, whereas most guidelines recommend serum 25OHD concentrations of >50 nmol/l for optimal bone health in older adults. However, the causal link between vitamin D and many extra-skeletal outcomes remains unclear. The VITAL, ViDA and D2d randomized clinical trials (combined number of participants >30,000) indicated that vitamin D supplementation of vitamin D-replete adults (baseline serum 25OHD >50 nmol/l) does not prevent cancer, cardiovascular events, falls or progression to type 2 diabetes mellitus. Post hoc analysis has suggested some extra-skeletal benefits for individuals with vitamin D deficiency. Over 60 Mendelian randomization studies, designed to minimize bias from confounding, have evaluated the consequences of lifelong genetically lowered serum 25OHD concentrations on various outcomes and most studies have found null effects. Four Mendelian randomization studies found an increased risk of multiple sclerosis in individuals with genetically lowered serum 25OHD concentrations. In conclusion, supplementation of vitamin D-replete individuals does not provide demonstrable health benefits. This conclusion does not contradict older guidelines that severe vitamin D deficiency should be prevented or corrected.