Next Article in Journal
Anti-Cancer Potential of Oxialis obtriangulata in Pancreatic Cancer Cell through Regulation of the ERK/Src/STAT3-Mediated Pathway
Previous Article in Journal
Changes in the Electrical Characteristics of Perovskite Solar Cells with Aging Time
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Effects of Vitamin D Supplementation on Inflammation, Colonic Cell Kinetics, and Microbiota in Colitis: A Review

School of Exercise and Nutritional Sciences, San Diego State University, San Diego, CA 92182, USA
*
Author to whom correspondence should be addressed.
Molecules 2020, 25(10), 2300; https://doi.org/10.3390/molecules25102300
Submission received: 16 April 2020 / Revised: 6 May 2020 / Accepted: 12 May 2020 / Published: 14 May 2020

Abstract

:
Vitamin D is widely known to regulate bone health, but there is increasing evidence that it may also ameliorate colitis through inflammation, cell proliferation and apoptosis, and the microbiota. The purpose of this review is to systematically examine the mechanisms by which vitamin D reduces colitis. PubMed and Web of Science were searched for articles published between 2008 and 2019 using key words such as “vitamin D,” “colitis,” “inflammatory bowel disease,” “inflammation,” “apoptosis,” “cell proliferation,” and “gut bacteria”. Retrieved articles were further narrowed and it was determined whether their title and abstracts contained terminology pertaining to vitamin D in relation to colitis in human clinical trials, animal studies, and cell culture/biopsy studies, as well as selecting the best match sorting option in relation to the research question. In total, 30 studies met the established criteria. Studies consistently reported results showing that vitamin D supplementation can downregulate inflammatory pathways of COX-2, TNF-α, NF-κB, and MAPK, modify cell kinetics, and alter gut microbiome, all of which contribute to an improved state of colitis. Although vitamin D and vitamin D analogs have demonstrated positive effects against colitis, more randomized, controlled human clinical trials are needed to determine the value of vitamin D as a therapeutic agent in the treatment of colitis.

1. Introduction

Vitamin D is a fat-soluble vitamin deemed essential for human health. Besides being known for maintaining bone homeostasis, Vitamin D has been implicated in innate and adaptive immunity, as well as having anti-inflammatory properties [1,2]. The Dietary Reference Intakes (DRIs) for vitamin D depend on age: 400 IU vitamin D/day for children <1 year old, 600 IU vitamin D/day for people ages 1–70 years old, and 800 IU vitamin D/day for people >70 years old [2,3]. Vitamin D is obtained through sunlight, dietary supplements, and vitamin D-rich foods such as cod liver oil, tuna, sardines, milk, eggs, certain mushrooms and fortified orange juice and dairy products [2]. Supplementation with vitamin D3 (5000 IU) improves serum concentrations of 25-hydroxy vitamin D (25-OHD), the precursor to the physiologically-active metabolite 1,25-duhydroxyvitamin D (1,25-OHD), in deficient (<30 ng/mL) individuals [4].
Colitis is a chronic, uncontrolled inflammation of the colon and destruction of the colonic crypt affecting the gastrointestinal tract; it causes malabsorption and diarrhea, and increased risk of developing colorectal adenocarcinoma [5,6,7,8,9]. In 2015, an estimated 3.1 million adults in the United States were diagnosed with either ulcerative colitis (UC) or Crohn’s disease; most were ≥45 years old, and non-Hispanic white (estimated 345,000 people) or Hispanic (estimated 395,000 people) [10]. According to the American Cancer Society, colorectal cancer is the third most common cancer diagnosed in the United States: 1 in 22 men, and 1 in 24 women [11]. History of UC for ≥8 years is one of the major risk factors for developing colorectal cancer [11,12]. Genetic, immune, environmental, and microbial factors may also play a role in the development of colitis [9,13,14].
Vitamin D status is negatively associated with inflammatory bowel disease (IBD) activity and clinical outcomes. A systematic review and meta-analysis of 27 studies found low vitamin D status, measured via 25-OHD, to be associated with increased disease activity, mucosal inflammation, and future clinical relapse, as well as decreased quality of life among IBD patients [15]. In subgroup analyses, these associations remained significant for patients with Crohn’s disease, but only associations for disease activity and future clinical relapse remained significant among patients with ulcerative colitis [15].
Many studies have demonstrated that vitamin D improves colitis in human and animal models. In two animal studies, vitamin D deficiency contributed to increased severity and mortality in experimental colitis and even in experimental colon cancer [7,8]. Mice fed increased dietary vitamin D had significantly lower pro-inflammatory cytokines associated with colon tumor growth [8]. It has been shown that proinflammatory cytokines, such as interleukin-6 (IL-6) and tumor necrosis alpha (TNF-α), inhibit vitamin D metabolism in colon cancer cells [16]. The one-time administration of 300,000 IU vitamin D3 reduced inflammation in UC subjects 90 days after intervention [17]. Associations between low serum vitamin D and the incidence of UC have been posited, but the exact mechanisms have not been well defined [7,9,17]. This review paper explores potential underlying mechanisms for the effects of vitamin D on colitis in terms of inflammation, colon cell kinetics, and the microbiota.

2. Results

2.1. Dose-Dependent Effects of Vitamin D Supplementation on Ulcerative Colitis and Crohn’s Disease

Supplementation of Vitamin D2 (ergocalciferol) and Vitamin D3 (cholecalciferol) improves serum vitamin D levels, with the latter having both higher potency and efficacy [18]. The optimal dose for Vitamin D supplementation in UC patients remains to be elucidated.
Vitamin D3 supplementation for 90 days at 4000 IU/day outperformed supplementation at 2000 IU/day in a prospective, double-blind, randomized trial of adults with hypovitaminosis D and UC [9]. Serum Vitamin D levels rose in both groups but to a larger degree in those receiving 4000 IU/day (16.8 ng/dL) than in those receiving 2000 IU/day (5.00 ng/dL). In the 4000 IU/day group, four of the ten patients achieved normal Vitamin D levels (>30 ng/mL) following supplementation compared to only one of the eight patients in the 2000 IU/day group. Quality of life measured via the Short IBD Questionnaire (SIBDQ) increased after 90 days to a statistically significant degree in the 4000 IU/day group, but not in the 2000 IU/day group [9].
In another double blind, randomized clinical trial, adults with hypovitaminosis D and mild to moderate UC were supplemented with either 1000 IU/day or 2000 IU/day of Vitamin D for 12 weeks. Measures included serum 25-hydroxy vitamin D (25-OHD), inflammatory bowel disease questionnaire-9 (IBDQ-9) scores, and a simple clinical colitis activity index questionnaire (SCCAI) scores. At the end of the study, supplementing with 2000 IU/day compared to 1000 IU/day resulted in a larger mean change in 25-OHD (6.7 ng/mL vs 0.2 ng/mL), larger improvement in mean IBDQ-9 score, and larger improvement in mean SCCAI scores [19].
A double blind, randomized, controlled trial compared the efficacy of two doses of weekly Vitamin D3 supplementation, 5000 IU and 10,000 IU/10 kg bodyweight/week for six weeks in pediatric IBD patients (N = 32). Serum 25-OHD was significantly higher compared to baseline in both groups at week 8 (24.0 to 41.5 ng/mL and 23.7 to 49.2 ng/mL, respectively), but only in the higher dose group at week 12 (30.8 ng/mL, p < 0.132 and 35.1 ng/mL, p < 0.001 respectively). No patient in either group experienced hypercalcemia or serious adverse events [20].

2.2. Vitamin D and Inflammation

2.2.1. Cyclooxygenase-2

Vitamin D has been shown to reduce inflammation and attenuate autoimmune disease by affecting both adaptive and innate immunity [21,22]. The anti-inflammatory properties of Vitamin D may work in part through the inhibition of the prostaglandin (PG) pathway. This inhibition is mediated by inhibiting the expression of cyclooxygenase-2 (COX-2), the enzyme responsible for synthesizing PGs, promoting the expression of 15-prostaglandin dehydrogenase (15-PGDH), the enzyme responsible for deactivating PGs, and disrupting PG signaling by decreasing EP and FP PG receptor expression [16,23,24] (Table 1). A human biopsy study examining COX-2 immunostaining among subjects with long-standing colitis revealed that over 90% of the biopsies from dysplastic lesions were positive for COX-2 expression [25]. The overexpression of COX-2 may accelerate tumor growth and the development of colitis to colorectal cancer through the induction of pro-oncogenic colonic crypt mutations [24,26]. Treatment of an adenocarcinoma colon cancer cell line, COGA-1A, with 10 nM 1,25-dihydroxyvitamin D (1,25-OHD) inhibited the upregulation of COX-2 mediated by TNF-α by 37% after 12 h [16]. Notably, 1,25-OHD has been found to inhibit COX-2 expression in a dose-dependent manner in murine macrophages through upregulation of thioesterase superfamily member 4 (THEM4) [27].
A Vitamin D analog, Ro26-2198, was found to inhibit COX-2 signaling and delay the onset of clinical colitis in an azoxymethane, dextran sulfate sodium mouse model, as well as to inhibit COX-2 induction and decrease proliferation in HCA-7 colon cancer cells. Ro26-2198 also inhibited increases in phosphoactive extracellular signal related kinase (ERK) and c-Myc intrinsic to the model [28].

2.2.2. CYP24A1 and CYP27B1

Activation of Vitamin D in the body is regulated by two genes, CYP27B1 and CYP24A1, with the former being responsible for activation and the latter for deactivation. Regulation of these enzymes is found to be important for colitis-associated colorectal cancer prevention by ensuring appropriate serum vitamin D to maintain normal cellular growth and differentiation [39]. The expression of these genes has been shown to be altered in the inflamed colonic tissue of IBD patients, resulting in a reduction of 25-OHD and impaired Vitamin D status [40]. Unsurprisingly, altered expression and polymorphisms of these genes, as well as low serum levels of 25-OHD, are associated with colorectal cancer [39,41].
A vitamin D-deficient, CYP27B1 knockout mouse model was shown to have shorter colon length, decreased body weight, thinner intestinal mucosa, and increased colonic crypt damage than wild-type counterparts [29]. The overexpression of CYP24A1 may play a role in the low serum 25-OHD levels seen in IBD patients, and modulating its expression may allow for better regulation of serum levels. In contrast, vitamin D analog BXL-62 showed significant reduction in proinflammatory cytokine expression such as TNF-α, and induced CYP24A1 expression in peripheral blood mononuclear cells in DSS-induced colitis in mice [30]. In the first known DSS-induced colitis mice study using gene therapy, CD11b+/Gr1+ monocytes were used as the cell vehicle as well as the macrophage-specific promoter (Mac1) to deliver and upregulate CYP27B1 expression in the colon to treat colitis [31]. It has been suggested that Gr1+ migrates to inflamed tissues first; however, Mac1 was only highly expressed in activated macrophages [31]. These activated macrophages were engineered to express the 1α-hydroxylase gene under the promoter of the Mac1 gene, so that CYP27B1 could be transported directly to the inflamed colon [31]. Mice injected with this treatment were found to have better survival, longer colon lengths, significantly reduced proinflammatory cytokines, and even mucosal regeneration compared to controls [31]. The upregulation of CYP24A1 following Vitamin D supplementation may be disadvantageous and should be considered when treating colitis [42].
Some studies have found that high CYP24A1 is prevalent in cancerous lesions in the colon compared to a normal colon. In a human biopsy study, CYP24A1 expression was 20 to 35 times higher and CYP27B1 was 4 times higher among low- and high-grade cancerous lesions in the colon compared to the normal colon [24]. Unbalanced, high basal levels of CYP24A1, however, may promote the progression of tumor growth, even in the colon [39]. How high the CYP24A1 expression has to be before tumorigenesis occurs remains unknown. Feedback mechanisms may be involved, but they are not well-defined at this time.
Single nucleotide polymorphisms (SNPs) are genetic variations that may potentially become biomarkers to predict risk of certain disease states [32]. Some studies explored CYP24A1 polymorphisms and risk for UC and colorectal cancer. One such study explored the association between CYP24A1 polymorphisms and risk of colonic polyps, colon cancer, and colitis in 144 subjects with colonic polyps, 96 subjects with colon cancer, and 44 subjects with UC [31]. SNPs identified and genotyped in CYP24A1 via an assay were rs4809957, rs6068816, rs6091822, and rs8124792 [36]. It was revealed that only SNPs rs4809957 and rs6068816 were significantly associated with risk of colonic polyps, colon cancer, and UC [31]. Similar studies also explored CYP24A1 polymorphisms for colorectal cancer risk, but risks for colitis were not discussed [43,44]. More research into the role of various Vitamin D activation and deactivation gene polymorphisms in UC treatment is warranted.

2.2.3. Tumor Necrosis Factor Alpha

TNF-α is a proinflammatory marker mediated by T-cells. It is one of the major pathogenic cytokines involved in the development of inflammatory bowel diseases, with elevated levels found in both CD and UC [16,45]. It has been shown that TNF-α promotes dysregulation of tight junction structures with remission following anti-TNF-α therapy [45,46,47]. If TNF-α-initiated inflammation is left unresolved, it may lead to colon cancer through angiogenesis and cell invasion [47,48]. Vitamin D may interfere with immune cells and suppress inflammatory cytokines, such as TNF-α, impeding intestinal inflammation [49].
A vitamin D analog, KH 1060, was shown to significantly increase serum vitamin D levels and decrease TNF-α levels among inflammatory bowel disease (IBD) patients. This analogue worked synergistically when combined with anti-TNF-α, resulting in lower TNF-α than either treatment alone [33]. Mice treated with vitamin D in a DSS-induced colitis model showed significantly reduced TNF-α expression compared to the control group. It was speculated that this reduction was due to a direct suppressive effect on cytokine expression or the maintenance of epithelial barrier function, thus averting innate immune system activation [34]. Similar results were seen in two other studies examining the effects of Vitamin D analogs, ZK191784 and TX527, on TNF-α expression in DSS-induced colitis murine models [35,36].

2.2.4. Nuclear Factor Kappa B

NF-κB is a key transcription factor that is crucial in various immune, physiological, and pathological responses; it is a major mediator of intestinal inflammation [50,51,52,53,54]. Among IBD patients, the expression of many inflammatory cytokines, such as NF-κB, IL-1 and IL-6, is increased in the intestinal mucosa [55]. In a bacterial-driven colitis and colon cancer murine model, the effects of two different Vitamin D doses were assessed: a maintenance dose of 1 IU/g of food and a high dose of 5 IU/g of food [8]. At 1 week postinfection, mice on the high-dose diet had decreased NF-κB activation in colonic epithelial cells. At 16 weeks, 41% of the mice on the maintenance-dose diet had cancer compared to only 11% of the mice on the high-dose diet. Decreased MAPK activation in the lamina propia leukocytes, inflammatory cell infiltrates, expression of proinflammatory cytokines, and IBD scores were also noted in the high-dose diet group [8].
Suppression of the NF-κB pathway and subsequent downstream expression of pro-inflammatory cytokines with 25-OHD has previously been confirmed in vitro [37]. A study on colon cancer-derived cells, HCT116, Caco-2, and SW480, treated with 20 nM 1,25-OHD for 24 h and followed by 100 ng/mL TNF-α for 12 h, revealed that vitamin D may maintain the intestinal epithelial barrier permeability by suppressing NF-κB activation [38].

2.2.5. Mitogen Activated Protein Kinase

The mitogen-activated protein kinase (MAPK) is a family of kinases that convert a variety of signals from the cell membrane to the nucleus in response to stimuli [56]. MAPK consists of three subunits, namely the extracellular signal-regulated kinase (ERK), c-Jun NH2-terminal kinase (JNK), as well as the p38 MAPK [56]. ERK is activated in response to growth stimuli, whereas JNK and p38 MAPK are activated in response to many cellular stresses such as inflammatory cytokines and DNA damage [56,57]. A subunit of p38 MAPK, p38α, was shown to regulate cellular response to stress and play a role in maintaining intestinal tissue homeostasis by cell differentiation [58,59]. Mice with downregulated p38α in intestinal epithelial cells in an AOM/DSS-colitis study were found to develop 29 low-grade tumors and 13 high-grade colonic tumors compared to 18 low-grade tumors and 7 high-grade tumors in wild-type mice [58]. Mice on 5 IU/g vitamin D showed a 2-fold decrease of JNK activation and 7-fold decrease of p38 activation in the colon compared to controls, suggesting that vitamin D may be used to treat IBD early on and prevent progression to colon cancer [8].

2.2.6. Retinoid X Receptor Alpha

RXRα belongs to the steroid nuclear receptor super family and is downregulated in murine colitis, colorectal cancer, and colon cancer cell lines, although the exact mechanism is yet to be identified [13,60]. RXRα+/− heterozygous mice treated with AOM/DSS showed significantly reduced RXRα expression and had a shorter colon length of 57.6 ± 9.4 mm compared to 67.8 ± 6.1 mm in the control group [60,61]. In the same study, heterozygous mice increased Snail1 and Snail2 expressions, which are transcription factors upregulated by inflammatory cytokines [60,61].
RXRα is also involved in the vitamin D signaling pathway via heterodimerization with VDR [60,62,63]. The biological activity of vitamin D may influence the immune system and inflammation by inducing VDR expression in the colon [6,9,14,64]. The VDR may play an anti-inflammatory role, especially in people with UC, as reduced VDR increases mucosal inflammation and barrier dysfunction [14]. In an experimental colitis mouse model, mice fed a vitamin D-deficient diet showed a lower serum vitamin D level of 20.1 ± 5.1 ng/mL, shortened colon length of 44.1 ± 4.6 mm, higher mortality rate at 7–10%, and lower VDR and RXRα expressions compared to control mice with a serum vitamin D level of 36.5 ± 4.7 ng/mL and colon length of 63.3 ± 3.8 mm [13].
Together with vitamin D and its proposed anti-inflammatory properties, vitamin D may reduce colitis by regulating COX-2, TNF-α, NF-κB through IkBa expression, RXR signaling, and vitamin D-activating enzymes CYP24A1 and CYP27B1.

2.3. Vitamin D and Cell Kinetics

2.3.1. Cell Proliferation

Normal colonic mucosa are made up of epithelial cells with folds called crypts, which are consistent throughout the colon [65,66]. Crypt renewal occurs at the bottom of the crypts where intestinal stem cells generate cell proliferation and differentiation, and thus replenish the epithelium [65,66,67]. In colitis, however, inflammation ultimately causes crypt disruption and architectural irregularities [65,66]. Vitamin D has been shown to promote mucosal regeneration through the modulation of signaling pathways, including VDR [65] (Table 2). Treatment of 10−8 M 1,25-OHD (calcitriol) in a cell study for 24 hr revealed increased expression of tight junction proteins, suggesting that vitamin D may play a role in maintaining the integrity of the tight junction complex [65]. VDR knockout mice showed impaired mucosal wound healing after DSS treatment, to the point where severe ulceration caused total crypt loss compared to wild-type mice [65].
Ki-67 is a nuclear protein constitutively expressed in mammalian cells, and is widely used as a cell proliferation marker in colitis and colon cancer [68]. DSS-treated rats had a significantly increased Ki-67 proliferation index compared to mice without DSS treatment [67]. Mice treated with vitamin D3 at 15, 30 and 60 IU⋅g−1⋅w−1 had significantly inhibited colonic cell proliferation through Ki-67 staining by 17%, 35%, and 41%, respectively, in an AOM/DSS-treated colitis model compared to controls [69]. Patients with colitis have significantly increased Ki-67 expression in low-grade dysplasia, high-grade dysplasia, and carcinoma compared to normal epithelium [70]. A longstanding UC biopsy study also revealed elevated Ki-67 expression in both low- and high-grade dysplasia [25].
Another biopsy study showed an inverse relationship between low serum vitamin D and Ki-67 expression among patients with precancerous and cancerous colon cells, as high proliferation is associated with cell malignancy [71]. Patients who had ≥50% Ki-67 expression in colon cells had a mean serum vitamin D level of 6.27 ng/mL; patients who had ≤50% Ki-67 expression had a mean serum vitamin D level of 13.42 ng/mL; and patients who had <15% Ki-67 expression and a mean serum vitamin D level of 20.53 ng/mL [71]. Treatment of 800 IU vitamin D/day for six months increased p21 expression by 242% compared to placebo [72]. A cyclin-dependent kinase inhibitor, p21 plays a critical role in cell differentiation, cell cycle control, apoptosis modulation, and tumorigenesis [72]. Vitamin D reduces colitis by promoting colonic mucosa regeneration and regulating cell proliferation via modulation of p21 expression.
Table 2. Characteristics of Published Clinical Trials Examining Vitamin D and Cell Kinetics.
Table 2. Characteristics of Published Clinical Trials Examining Vitamin D and Cell Kinetics.
Author (Year)Study Type, PopulationMeasurementsResultsConclusion
Kong et al. (2008) [65]VDR+/+ (wild-type) and VDR−/− (KO) mice
2.5% DSS treatment; cell study
Colonic samples; histology, immunostaining, electron microscopy, Western blotting, RT-PCRVDR KO mice had more severe colitis/ulceration, symptoms like diarrhea, gross bleeding, rectal prolapse, ↑ mucosal permeability, impaired epithelial junction compared to wild-type mice; vitamin D treatment ↑ tight junction proteins in 24 hVitamin D may protect intestinal epithelial barrier by increasing tight junction proteins
Xin et al. (2017) [69]C57BL/6 mice
12.5 mg/kg AOM, 2.5% DSS treatment;
Cell culture (SW480 cells) w/ 100 nmol/L 1,25-OHD treatment
Serum vitamin D, calcium, creatinine levels; vitamin D (15, 30, or 60 IU⋅g−1⋅w−1); transient transfection, luciferase assay; RNA isolation, RT-PCR, immunoprecipitation and Western blottingAOM/DSS treatment ↓ serum vitamin D levels; vitamin D administration significantly ↓ the number and colorectal neoplasms in a dose-dependent manner by up to 59%, ameliorated colon length shortening, ↓ cell proliferation in the colon
↓ β-catenin expression in vivo, ↑β-catenin binding to E-cadherin in vivo
Vitamin D is beneficial against AOM-DSS-induced colitis-associated carcinogenesis
E-cadherin upregulation is beneficial on vitamin D’s preventive effect on β-catenin
Fedirko et al. (2009) [72]Randomized, double-blind, placebo-controlled, clinical trial in men and women (N = 92) with confirmed colorectal adenomaAutomated immunohistochemistry and image analysis to detect and quantify expression and distribution of p21waf1/cip1, MIB-1, and hTERT in colorectal cryptsp21 expression ↑ 2-fold in Vitamin D and Vitamin D plus calcium but not in placebo group; no change in MIB-1 or hTERT expression; proportion of hTERT extending into upper 40% of the crypts ↓ by 15%Calcium and Vitamin D promote differentiation of colorectal epithelial cells and may regulate colorectal crypt proliferative zone in patients with sporadic adenoma
Zhu et al. (2015) [73]Biopsies from IBD patients;
C57BL6/J mice treated with 100 mg/kg TNBS and randomly divided into 2 groups: TNBS and control w/ either paricalcitol 0.5 µg/kg body weight or dissolvent only
Western blotting, hematoxylin/eosin staining, serum vitamin D levels, FTC-dextran intestinal permeability↓ serum vitamin D and VDR among IBD patients; paricalcitol ↓ TNBS-induced colitis (i.e., minor weight loss, mild colitis, minimal histological damage), protects intestinal permeability, inhibits intestinal epithelial cell apoptosis by ↓ PUMA expressionVitamin D and vitamin D analogs show promise when treating IBD by downregulating PUMA expression

2.3.2. E-Cadherin

E-cadherin is a transmembrane linker protein and calcium-dependent cell adhesion molecule for colon crypt structure and function, making it important for mediating and maintaining intestinal homeostasis and epithelial architecture [74]. The loss of E-cadherin expression transitions adenoma to carcinoma, is a hallmark of colon carcinogenesis, and is prevalent in colitis [75,76]. The deletion of E-cadherin in mice resulted in severe loss of body weight, severe loss of intestinal epithelial structure, an increased number of apoptotic cells, impaired bacterial defense, and length reduction of the small intestine compared to controls [76]. In an AOM/DSS-colitis mice study, vitamin D3 supplementation (15, 30, and 60 IU·g−1·w−1 3x/week) showed significant preventive effects in high doses, decreased the number of neoplasms by 33–63%, and increased E-cadherin expression, suggesting inhibition of colonic carcinogenesis progression [69]. E-cadherin in epithelial and immune cells was found to be expressed less in CYP27B1 knockout mice compared to their wild type counterparts in this DSS-induced colitis study [77]. These studies suggest that vitamin D improves colitis and prevents colon cancer progression by enhancing E-cadherin expression.

2.3.3. Wnt/β-Catenin

Wnt signaling is crucially involved in stem cell maintenance and epithelial cell proliferation [78,79,80]. Wnt is activated in most colon cancers; therefore, it can be used as a potential biomarker for colon health [78,81,82]. Wnt may also contribute to carcinogenesis from colitis to colon cancer [78,81,82]. Wnt is capable of signaling in a paracrine and autocrine manner, and the absence of Wnt promotes the degradation of β-catenin [79]. β-catenin is a key component of the Wnt pathway, and the activation of β-catenin predisposes humans and mice to both colitis and colon cancer [78,79,82,83]. In a biopsy study, 100% of the patients in the colitis-associated colorectal cancer group demonstrated β-catenin expression compared to none of the patients in the control group [81]. In another biopsy study, there were significantly more colonic Wnt and β-catenin stained cells in colitis patients (2.5-fold) and colorectal cancer patients (4.5-fold increase) compared to colon tissue from healthy controls [82]. Tumor samples extracted from patients with chronic UC and colon cancer were found to have elevated β-catenin expression after staining compared to noncancerous colonic tissues [83].
Supplementation of vitamin D3 (15, 30, and 60 IU·g−1·w−1 3x/week) in an AOM/DSS-colitis mouse model revealed decreased β-catenin, number and burden of colorectal neoplasms, and shortening of colon length, all in a dose-dependent manner [69]. Many other studies have explored the effects of vitamin D on the Wnt signaling pathway in the colon, but few have done so specifically in colitis [84,85].

2.3.4. Cell Apoptosis

Apoptosis is a process of natural cell death by eliminating genetically-mutated cells [86,87,88]. Homeostasis of the colonic epithelium requires a balance among cell proliferation, necrosis, and apoptosis [86,87,88,89]. Excessive intestinal epithelial cell apoptosis is a major cause of loss of epithelial cells and mucosal permeability, contributing to the development of colitis [7,90,91]. In a DSS-induced colitis rat model, the epithelial apoptotic index was significantly increased, at 7.2% ± 1.2% compared to 2.8% ± 1.2% in the control group [90]. Increased apoptosis in the DSS-treated colitis model showed a significantly decreased number of intact colonic crypts [67]
VDR signaling has been shown to prevent apoptosis by downregulating p53-upregulated modulator of apoptosis (PUMA) by blocking NF-κB in TNBS-induced colitis [91]. A tumor suppressing gene, p53 is crucial for cell fate and prevents the spread of damaged cells [91]. Along with p53, PUMA is a potent inducer of apoptosis that is correlated with the severity of colitis, and is crucial to mediating intestinal epithelial cell apoptosis [7,92]. In an experimental colitis mouse model, VDR knockout mice developed more severe colitis with significant colonic epithelial cell apoptosis compared to control mice [91]. In another study, VDR knockout mice had significantly increased apoptotic epithelial cells compared to wild-type mice [7]. Mice treated with a vitamin D analog, paricalcitol, in a 2,4,6-trinitrobenzene sulfonic acid (TNBS) induced colitis model, had decreased severity of colitis compared to controls mediated in part through an inhibition of intestinal epithelial cell (IEC) apoptosis from a downregulation of PUMA expression [73]. The absence of PUMA was found to relieve DSS- and TNBS-induced colitis, as well as to inhibit IEC apoptosis in PUMA-knockout mice compared to wildtype mice [92]. Human biopsies have revealed elevated levels of p53 and PUMA in the inflamed mucosal tissues of UC patients compared to those of healthy controls [93]. Vitamin D protects against colitis through the prevention of apoptosis via downregulation of PUMA.

2.4. Vitamin D and Microbiota

The gastrointestinal tract houses a variety of microbiota that are important for nutrient absorption, immunity, and intestinal epithelial barrier homeostasis [94,95]. Dysbiosis or abnormal interactions between the host and the gut microbiota lead to gastrointestinal diseases such as colitis [94,95,96,97]. UC patients have an unstable microbiota profile and reduced bacterial species richness compared to healthy patients [98]. Probiotics aid in intestinal homeostasis, and studies have shown potential therapeutic benefits of probiotics in colitis [94,95,96]. Probiotics have been shown to enhance VDR transcriptional activity, induce remission, reduce clinical activity scores, and prevent pouchitis, a complication in colitis following ileal pouch-anal anastomosis surgery [94,95,96]. In an experimental Salmonella colitis mouse model, probiotics Lactobacillus rhamnosus strain GG and Lactobacillus plantarum increased VDR expression and signaling, suggesting that VDR is necessary to prevent pathogenic invasion in the colon [96] (Table 3).
Serum vitamin D levels are associated with the function and composition of the intestinal microbiome [99,100]. Clostridium difficile (C. difficile) is a Gram-positive, anaerobic, spore-forming bacillus associated with diarrhea, sepsis, megacolon, and pseudomembranous colitis [101]. C. difficile is associated with a four-fold increase in mortality, increased hospitalization, and risk for colectomy up to 5 years after the first episode [102]. C. difficile-induced colitis is a costly and common healthcare-associated infection following antibiotic treatment, causing approximately 29,000 annual deaths in the United States [103]. C. difficile infection (CDI) is correlated with low serum vitamin D in IBD patients (mean of 20.4 ng/mL) versus non-CDI IBD patients (mean of 27.1 ng/mL), with a 4% reduction in CDI risk associated with each 1 ng/mL increase in plasma 25-OHD [102]. One innate line of defense against the toxic effects of C. difficile is cathelicidin, an anti-inflammatory, antimicrobial peptide activated by plasma 25-OHD [98,102,104]. Cathelicidin levels are higher in UC patients than healthy controls, but decrease with disease severity [98,102,104] A randomized, controlled trial in UC patients found a 3-fold increase in the cathelicidin precursor, human cationic antimicrobial protein 18 (hCAP18), gene expression following a single intramuscular injection containing 300,000 IU Vitamin D3 [17]. Serum levels of 25-OHD are positively associated with serum and colonic cathelicidin among UC patients. Serum cathelicidin is negatively associated with histologic inflammation, and clinical relapse risk though these associations did not remain significant in multivariate analyses. In these multivariate analyses, only 25-OHD levels ≤35 ng/mL were independently associated with an increase in histologic inflammation and baseline histologic inflammation independently with an increase in clinical relapse risk [105].
It has been suggested that VDR acts as a master regulator of intestinal inflammation by regulating both autophagy and antimicrobial peptide production [106]. VDR KO mice have a higher susceptibility to DSS-induced colitis and subsequent dysbiosis with fewer butyrate-producing bacteria and increased E. coli and Bacteriocides [106]. Decreased levels of butyrate-producing bacteria and butyrate itself have been noted in UC patients relative to healthy controls [107]. The pretreatment of human IEC with butyrate was found to increase VDR expression in a dose-dependent manner [106].
Angiogenin-4 (Ang4) is an antimicrobial protein involved in host containment of enteric bacteria [108]. The expression of Ang4 is downregulated in the colon after DSS treatment, and even lower in vitamin D-deficient mice receiving either tap water or DSS [108]. Decreased Ang4 expression in vitamin D-deficient mice leads to homeostasis disturbance in the colon compared to vitamin D sufficient mice [108]. It is possible that vitamin D may regulate enteric bacteria by promoting Ang4 expression, but the mechanisms have not yet been identified [108].
Colonic tissue samples from IBD patients had phylogenetic distribution from 4 phyla of bacteria: Firmicutes (49%), Bacteroidetes (23%), Proteobacteria (21%), and Actinobacteria (5%) [109]. A stool sample study revealed that Firmicutes (60%), Verrumicrobiota (25%), and Bacteroidetes (13%) were the main phyla from an IBD patient, whereas Firmicutes (59%), Bacteroidetes (28%), and Actinobacteria (13%) were found from a colon carcinoma patient [110].
The administration of vitamin D to patients with UC has been shown to reduce intestinal inflammation. These reductions coincided with increases in Enterobacteriaceae and a trend of reduction of the mucolytic species Ruminococcus gnavus, but not with changes in the overall fecal microbial diversity [98]. The supplementation of vitamin D in individuals with Crohn’s disease has also been shown to influence microbial communities. One week of supplementation increased the abundance of Alistipes (Bacteroidetes); however, by the second week prevalence of Bacteroidetes decreased, while Blautia, Firmicutes, Fusicatenibacter, Intestinibacter, and Veillonella increased. By the third week Parabacteroides were at their highest. By week 4, Lactobacillus and Megasphera were at their highest. From week 0 to week 4 of vitamin D supplementation, the number of bacterial taxa decreased [111].
There are bacterial changes observed in feces between Cyp KO mice and Cyp wild-type mice [77]. Cyp KO and VDR KO mice had more Bacteroidetes and Proteobacteria phyla, but fewer Firmicutes and Deferribacteres phyla than wild-type mice [77]. However, vitamin D treatment reduced the numbers of the Helicobacteraceae family from the Proteobacteria phylum in Cyp KO mice [77]. Vitamin D protects against colitis through VDR by regulating the gut microbiota and decreasing pathogenic infections in the colon.
Table 3. Characteristics of Published Clinical Trials Examining Vitamin D and the Microbiota.
Table 3. Characteristics of Published Clinical Trials Examining Vitamin D and the Microbiota.
Author (Year)Study Type, PopulationMeasurementsResultsConclusion
Sharifi et al. (2016) [17]Double-blind, randomized, controlled trial with parallel design including UC patients in remission (N = 90)Serum levels of 25-OHD, PTH, Calcium, ESR, and hs-CRP and cathelicidin expression via qRT-PCR pre and post 90 days of 300,000 IU intramuscular Vitamin D or saline placeboFrom pre to post, ↑ 25 -OH D, ↑ hCAP18 (human cationic antimicrobial protein 18) gene expression, ↓ Hs-CRP and ↓ ESR only in Vitamin D groupSupplementation of Vitamin D may benefit UC patients as evidenced by ↓ ESR, ↓ hs-CRP, and ↑ LL37 gene expression
Wu et al. (2015) [96]C57BL/6 mice and VDR KO C57BL/6 mice;
Salmonella-induced colitis mouse model
Salmonella infection
Histology, Western blotting; immunofluorescence; VDR protein expression transcriptional activity; RT-PCR
VDR KO mice significantly lost more body weight, probiotics had no protection from Salmonella-induced colitis, and more Salmonella infection occurred compared to VDR+/+ mice; Lactobacillus rhamnosus strain GG (LGG) and Lactobacillus plantarum (LP) ↑ VDR expression in vitroProbiotics are suggested to enhance VDR expression and may help protect against colitis
Ananthakrishnan et al. (2014) [102]Multi-institutional cohort of IBD patients (n = 3188); 35 patients developed Clostridium difficile infection (CDI) w/ mean age of 60.5 years oldSerum vitamin D level measurement via radioimmunoassay, high-performance liquid chromatography w/ mass spectrometryIBD patients with CDI who died had a mean serum average of 12.8 + 8.1 ng/mL compared to IBD patients who remained alive at the end of follow up (24.3 + 13.2 ng/mL); 1 ng/mL increase in serum vitamin D = w/ a 4% CDI reductionHigher serum vitamin D levels were associated with a decreased CDI risk
Wu et al. (2015) [106]Conditional VDR KO mouse model (VDRΔIEC)
5% DSS treatment, butyrate treatment (fermentation product of gut microbes)
Vitamin D-responsive element transcriptional activity, Western blotting, intestine histology, immunofluorescence, lysotracker staining, RT-PCR, chromatin immunoprecipitation (CHIP) assayVDRΔIEC susceptible to DSS-induced colitis, ↓ weight, ↓ cecum length, fecal blood present, ↑ intestinal inflammation, fewer butyrate-producing bacteria and butyrate compared to no DSS treatment; butyrate ↑ VDR expression and inhibited inflammation; ↑ E. coli and BacteriocidesVDR may help regulate intestinal homeostasis via production of antimicrobial peptides
Lagishetty et al. (2010) [108]C57BL/6 mice raised on a normal diet (n = 16) or vitamin D-deficient diet (n = 16) × 6 weeks
2.5% DSS treatment
Tissue collection/analysis, clinical colitis score, histological colitis score, flow cytometry, RT-PCR; Ang4 immunohistochemical analysisVitamin D-deficient mice treated with DSS had more severe colitis, ↓ colonic Ang4 expression, ↑ bacterial infiltration compared to mice with normal diet; DSS-treated vitamin D-deficient mice had ↓ serum vitamin D level (2.5 ± 0.1 ng/mL) compared to mice with normal diet (24.4 ± 1.8 ng/mL)Ang4 promotes bacterial innate immunity against gut microbes and its function under a low serum vitamin D levels is impeded
Ooi et al. (2013) [76]Cyp wild-type mice, Cyp KO mice, VDR wild-type mice, VDR KO mice
3.5% DSS treatment x 5 days
Fecal samples from mice
Denaturing gradient gel electrophoresis, metagenomic analysis, RT-PCR, cell isolation
Firmicutes and Bacteroidetes are the dominant phyla in all mice; Cyp KO and VDR KO had ↓ Firmicutes and Deferribacteres, ↑ Proteobacteria and Bacteroidetes phyla than wild-type mice; vitamin D treatment ↓ HelicobacteraceaeVitamin D and VDR may influence microbiome composition and protect against GI insults
Garg et al. (2018) [98]Patients with active UC, inactive UC and noninflammatory bowel disease controls; received 40,000 units cholecalciferol weekly for 8 weeks.Markers of inflammation and fecal microbiotaPatients with active UC ↓ faecal calprotectin levels; this did not change in patients with inactive UC or non-IBD controls. No changes in overall fecal bacterial diversity. were noted although a significant ↑ in Enterobacteriaceae abundance in patients with UCVitamin D supplementation reduced intestinal inflammation in patients with active UC, with an increase in Enterobacteriaceae and a trend to reduction in the mucolytic species Ruminococcus gnavus but no change in overall fecal microbial diversity
Schaffler et al. (2018) [111]A prospective, longitudinal, controlled interventional analysis in seven patients with Crohn’s disease (CD) in clinical remission and 10 healthy controls (HC); orally administration of vitamin DIntestinal bacterial composition↓ bacterial richness in the CD microbiome.
Alistipes, Barnesiella, unclassified Porphyromonadaceae (both Actinobacteria), Roseburia, Anaerotruncus, Subdoligranulum and an unclassified Ruminococaceae (all Firmicutes)
Vitamin D has a specific influence on the bacterial communities in CD, but not in HC. Administration of vitamin D may have a positive effect in CD by modulating the intestinal bacterial composition and also by increasing the abundance of potential beneficial bacterial strains. Vitamin D did not change the bacterial communities in HC.

3. Discussion

Evidence from tissue biopsies, cell line studies, murine models, and clinical human trials suggest that vitamin D may be beneficial against colitis by reducing inflammation, inhibiting cell proliferation and apoptosis, and regulating the gut microbiota. Furthermore, these benefits are often exerted in a dose-dependent manner with no clearly established ceiling. The reduction in inflammation appears to be multifactorial, resulting from a modification of the expression of COX-2, CYP24A1 and CYP27B1, MAPK, and RXRα, and a reduction of cytokines including TNF-α and NF-kB. A decrease in cell proliferation and β-catenin and increase in cell apoptosis and E-cadherin expression also helps explain the beneficial effect of Vitamin D. Pathogenic alterations in the composition of gut microbiota associated with colitis may be mitigated by Vitamin D supplementation through increased VDR signaling and Ang4 expression. Most current evidence supporting the benefit of Vitamin D supplementations to colitis stems from cell line studies and murine models. While the evidence from human-based genetic studies and tissue biopsies suggest similar underlying mechanisms, the success of Vitamin D in treating colitis in murine models needs to be confirmed in human clinical trials. Furthermore, while Vitamin D analogs can be effective in certain models, evidence of their efficacy in humans is lacking. Mechanistic evidence from animal and cell models, such as that cited within this review, should be considered when developing much needed human clinical trials.
The beneficial effects of vitamin D on IBD also appear to be mediated through actions on inflammatory cytokines including TNF- α, IL-6, and IL-12 and anti-inflammatory cytokines including IL-4, IL-10, and TGB- β. The ratio of these anti-inflammatory to inflammatory cytokines is improved by restoring vitamin D status via supplementation in IBD patients who otherwise often have insufficient levels [112].
According to our findings, VDR plays a critical role in the action of vitamin D against colitis. Inflammation-related signaling including E-cadherin, RXRα, and wnt/β-catenin seems to work via VDR. VDR KO mice generally suffer from impaired mucosal healing, severe crypt loss, and higher apoptotic ratio. These studies indicate that VDR status is crucial and appears to be some sort of defense mechanism in colitis. Colitis decreases cell proliferation and increases apoptosis; a lack of VDR in several mice studies was shown to affect cell proliferation and apoptosis. VDR is involved in maintaining the tight junction and intestinal epithelial barriers that regulate cell proliferation.
The role of the microbiome is very important in colitis prevention or treatment. However, more studies are needed on the potential mechanisms of vitamin D supplementation on the microbiota in IBD patients.
Due to the short half-life and lower circulating levels of the active form, 1,25-OHD, its precursor, 25-OHD, is often used to assess the vitamin D status in the body [113]. However, in order for 25-OHD to accurately reflect 1,25-OHD levels, the 1α-hydroxylating enzyme CYP27B1, which converts the former to the latter, would need to be optimally expressed [29]. Similarly, optimal levels of the catabolizing hydroxylase CYP24A1, which degrades 1,25-OHD, would be essential. CYP24A1 is induced mainly by 1,25-OHD, and CYP24A1 reduces elevated local 1,25-OHD levels in a negative feedback manner [39]. In those with colitis, the ratio of CYP27B1 to CYP24A1 may prove to be important for successful treatment [39].
In addition, CYP27B1 polymorphisms can lead to Vitamin D insufficiency and supplementation of Vitamin D increases CYP24A1 expression. Some studies have shown that high CYP24A1 can induce colon carcinogenesis, but no current studies have indicated how high it would have to be to do so. The role of CYP27B1 and CYP24A1 in regulating serum Vitamin D levels is both complex and consequential. Future studies should include the dose effects of vitamin D on CYP24A1 expression to determine the threshold at which CYP24A1 promotes colon carcinogenesis.
It was observed that the criteria for defining Vitamin D statuses varied between studies [9,17,29,38,99]. While the Institute of Medicine has specified serum vitamin D thresholds indicative of insufficiency and deficiency with regards to bone health [114], levels which optimize human immune function remain unclear and require further investigation.
Vitamin D analogs have shown promising results, and their utility in treating colitis should continue to be explored. Administration of 1,24(OH)2D2 was found to have similar effects compared to 1,25(OH)2D2 on reducing colitis symptoms, but 1,24(OH)2D2 presents less risk in raising serum calcium levels [63]. Having high serum calcium, or hypercalcemia, is a potentially life-threatening condition that can cause progressive kidney failure, dehydration, and granulomatous diseases such as tuberculosis and sarcoidosis [106]. The relationship between calcium and vitamin D, specifically concerning the various types of vitamin D, should be further investigated as potential therapeutic options for colitis.
Animal studies remain popular, as genetic techniques can optimize disease models to better understand the pathology of ulcerative colitis. There are limited human clinical trials on the effect of vitamin D on colitis. More randomized, controlled human clinical trials are needed. In conclusion, vitamin D may benefit sufferers of colitis by reducing inflammatory cytokines, maintaining intestinal epithelial cell integrity by modulation of cell proliferation and apoptosis, and reducing pathogenic infections.

4. Materials and Methods

PubMed and Web of Science were the primary databases used to search for articles—published between 2008 and 2019—using key words such as “vitamin D and colitis,” “vitamin D and inflammatory bowel disease,” “inflammation,” “apoptosis,” “cell proliferation,” and “gut bacteria”. PubMed initially yielded 447 articles that were then evaluated further. Secondary inclusion criteria were based on whether or not titles and abstracts included “vitamin D” in relation to colitis in human subjects, animal models, and cell culture studies, as well as selecting the best match sorting option on PubMed according to key words. Upon further evaluation, 251 articles met the secondary criteria. After further checking based on the criteria of original studies and mechanism-based vitamin D intervention on colitis, 30 articles were finally chosen that were most relevant to vitamin D and colitis on inflammation (Table 1), cell kinetics (Table 2), and the microbiota (Table 3).

Author Contributions

Conceptualization, methodology, P.M.G. and M.Y.H.; formal analysis and original draft preparation, P.M.G.; investigation and data curation, P.M.G. and J.M.; review and editing, J.M., M.Y.H. and D.K.; supervision, M.Y.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding. The APC was funded by MDPI.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

25-OHD25-hydroxy vitamin D
1,25-OHD1,25-duhydroxyvitamin D
AOMazoxymethane
BMIbody mass index
CDCCenters for Disease Control and Prevention
CDIClostridium difficile infection
COX-2cyclooxygenase-2
CRPC-reactive protein
DRIDietary Reference Intake
DSSdextran sulfate sodium
ERKextracellular signal related kinase
ESRerythrocyte sedimentation rate
hTERTTelomerase reverse transcriptase
IBDinflammatory bowel disease
IL-6interleukin-6
IUinternational units
MLCKmyosin light chain kinase pathway
LL37cathelicidin
MAPKmitogen-activated protein kinase
PUMAp53-upregulated modulator of apoptosis
RXRαRetinoid X Receptor alpha
TNBS2,4,6-trinitrobenzene sulfonic acid
TNF-αtumor necrosis factor-alpha
UCulcerative colitis
USDHHSUnited States Department of Health and Human Services
VDRvitamin D receptor

References

  1. Vukić, M.; Neme, A.; Seuter, S.; Saksa, N.; De Mello, V.D.F.; Nurmi, T.; Uusitupa, M.; Tuomainen, T.-P.; Virtanen, J.K.; Carlberg, C. Relevance of vitamin D receptor target genes for monitoring the vitamin D responsiveness of primary human cells. PLoS ONE 2015, 10, e0124339. [Google Scholar] [CrossRef] [Green Version]
  2. Office of Dietary Supplements-Vitamin D. Available online: https://ods.od.nih.gov/factsheets/VitaminD-HealthProfessional/ (accessed on 13 March 2020).
  3. Institute of Medicine (US). Committee to Review Dietary Reference Intakes for Vitamin D and Calcium. In Dietary Reference Intakes for Calcium and Vitamin D; The National Academies Collection: Reports funded by National Institutes of Health; Ross, A.C., Taylor, C.L., Yaktine, A.L., Del Valle, H.B., Eds.; National Academies Press (US): Washington, DC, USA, 2011. [Google Scholar]
  4. Lukaszuk, J.M.; Luebbers, P.E. 25(OH)D status: Effect of D3 supplement. Obes. Sci. Pract. 2017, 3, 99–105. [Google Scholar] [CrossRef] [PubMed]
  5. Agoff, S.N.; Brentnall, T.A.; Crispin, D.A.; Taylor, S.L.; Raaka, S.; Haggitt, R.C.; Reed, M.W.; Afonina, I.A.; Rabinovitch, P.S.; Stevens, A.C.; et al. The role of cyclooxygenase 2 in ulcerative colitis-associated neoplasia. Am. J. Pathol. 2000, 157, 737–745. [Google Scholar] [CrossRef] [Green Version]
  6. Wada, K.; Tanaka, H.; Maeda, K.; Inoue, T.; Noda, E.; Amano, R.; Kubo, N.; Muguruma, K.; Yamada, N.; Yashiro, M.; et al. Vitamin D receptor expression is associated with colon cancer in ulcerative colitis. Oncol. Rep. 2009, 22, 1021–1025. [Google Scholar] [CrossRef] [Green Version]
  7. Liu, W.; Chen, Y.; Golan, M.A.; Annunziata, M.L.; Du, J.; Dougherty, U.; Kong, J.; Musch, M.; Huang, Y.; Pekow, J.; et al. Intestinal epithelial vitamin D receptor signaling inhibits experimental colitis. J. Clin. Investig. 2013, 123, 3983–3996. [Google Scholar] [CrossRef] [Green Version]
  8. Meeker, S.; Seamons, A.; Paik, J.; Treuting, P.M.; Brabb, T.; Grady, W.M.; Maggio-Price, L. Increased dietary vitamin D suppresses MAPK signaling, colitis, and colon cancer. Cancer Res. 2014, 74, 4398–4408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Mathur, J.; Naing, S.; Mills, P.; Limsui, D. A randomized clinical trial of vitamin D3 (cholecalciferol) in ulcerative colitis patients with hypovitaminosis D3. PeerJ 2017, 5, e3654. [Google Scholar] [CrossRef]
  10. Dahlhamer, J.M.; Zammitti, E.P.; Ward, B.W.; Wheaton, A.G.; Croft, J.B. Prevalence of Inflammatory Bowel Disease Among Adults Aged ≥18 Years — United States, 2015. MMWR Morb. Mortal. Wkly. Rep. 2016, 65, 1166–1169. [Google Scholar] [CrossRef] [Green Version]
  11. Key Statistics for Colorectal Cancer. Available online: https://www.cancer.org/cancer/colon-rectal-cancer/about/key-statistics.html (accessed on 13 March 2020).
  12. Colorectal Cancer Prevention (PDQ®)–Health Professional Version. Available online: https://www.cancer.gov/types/colorectal/hp/colorectal-prevention-pdq (accessed on 13 March 2020).
  13. Knackstedt, R.W.; Moseley, V.R.; Sun, S.; Wargovich, M.J. Vitamin D receptor and retinoid X receptor α status and vitamin D insufficiency in models of murine colitis. Cancer Prev. Res. 2013, 6, 585–593. [Google Scholar] [CrossRef] [Green Version]
  14. Li, Y.C.; Chen, Y.; Du, J. Critical roles of intestinal epithelial vitamin D receptor signaling in controlling gut mucosal inflammation. J. Steroid Biochem. Mol. Biol. 2015, 148, 179–183. [Google Scholar] [CrossRef] [Green Version]
  15. Gubatan, J.; Chou, N.D.; Nielsen, O.H.; Moss, A.C. Systematic review with meta-analysis: Association of vitamin D status with clinical outcomes in adult patients with inflammatory bowel disease. Aliment. Pharmacol. Ther. 2019, 50, 1146–1158. [Google Scholar] [CrossRef]
  16. Hummel, D.M.; Fetahu, I.S.; Gröschel, C.; Manhardt, T.; Kállay, E. Role of proinflammatory cytokines on expression of vitamin D metabolism and target genes in colon cancer cells. J. Steroid Biochem. Mol. Biol. 2014, 144 Pt A, 91–95. [Google Scholar] [CrossRef] [Green Version]
  17. Sharifi, A.; Hosseinzadeh-Attar, M.J.; Vahedi, H.; Nedjat, S. A randomized controlled trial on the effect of vitamin D3 on inflammation and cathelicidin gene expression in ulcerative colitis patients. Saudi J. Gastroenterol. 2016, 22, 316–323. [Google Scholar] [CrossRef] [PubMed]
  18. Tripkovic, L.; Lambert, H.; Hart, K.; Smith, C.P.; Bucca, G.; Penson, S.; Chope, G.; Hyppönen, E.; Berry, J.; Vieth, R.; et al. Comparison of vitamin D2 and vitamin D3 supplementation in raising serum 25-hydroxyvitamin D status: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2012, 95, 1357–1364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Karimi, S.; Tabataba-vakili, S.; Yari, Z.; Alborzi, F.; Hedayati, M.; Ebrahimi-Daryani, N.; Hekmatdoost, A. The effects of two vitamin D regimens on ulcerative colitis activity index, quality of life and oxidant/anti-oxidant status. Nutr. J. 2019, 18, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Simek, R.Z.; Prince, J.; Syed, S.; Sauer, C.G.; Martineau, B.; Hofmekler, T.; Freeman, A.J.; Kumar, A.; McElhanon, B.O.; Schoen, B.T.; et al. Pilot study evaluating efficacy of 2 regimens for hypovitaminosis D repletion in pediatric inflammatory bowel disease. J. Pediatr. Gastroenterol. Nutr. 2016, 62, 252–258. [Google Scholar] [CrossRef] [Green Version]
  21. Liu, N.; Nguyen, L.; Chun, R.F.; Lagishetty, V.; Ren, S.; Wu, S.; Hollis, B.; DeLuca, H.F.; Adams, J.S.; Hewison, M. Altered endocrine and autocrine metabolism of vitamin D in a mouse model of gastrointestinal inflammation. Endocrinology 2008, 149, 4799–4808. [Google Scholar] [CrossRef] [Green Version]
  22. Adams, J.S.; Hewison, M. Unexpected actions of vitamin D: New perspectives on the regulation of innate and adaptive immunity. Nat. Clin. Pract. Endocrinol. Metab. 2008, 4, 80–90. [Google Scholar] [CrossRef] [Green Version]
  23. Krishnan, A.V.; Moreno, J.; Nonn, L.; Malloy, P.; Swami, S.; Peng, L.; Peehl, D.M.; Feldman, D. Novel pathways that contribute to the anti-proliferative and chemopreventive activities of calcitriol in prostate cancer. J. Steroid Biochem. Mol. Biol. 2007, 103, 694–702. [Google Scholar] [CrossRef]
  24. Brozek, W.; Manhardt, T.; Kállay, E.; Peterlik, M.; Cross, H.S. Relative expression of Vitamin D hydroxylases, CYP27B1 and CYP24A1, and of cyclooxygenase-2 and heterogeneity of human colorectal cancer in relation to age, gender, tumor location, and malignancy: Results from factor and cluster analysis. Cancers 2012, 4, 763–776. [Google Scholar] [CrossRef]
  25. Frăţilă, O.C.; Iliaş, T.I. COX-2 and Ki-67 immunohistochemical markers in the assessment of long-standing ulcerative colitis associated dysplasia. Rom. J. Morphol. Embryol. 2013, 54, 143–149. [Google Scholar] [PubMed]
  26. Leedham, S.J.; Graham, T.A.; Oukrif, D.; McDonald, S.A.C.; Rodriguez-Justo, M.; Harrison, R.F.; Shepherd, N.A.; Novelli, M.R.; Jankowski, J.A.Z.; Wright, N.A. Clonality, founder mutations, and field cancerization in human ulcerative colitis-associated neoplasia. Gastroenterology 2009, 136, 542–550.e6. [Google Scholar] [CrossRef] [PubMed]
  27. 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. J. Biol. Chem. 2014, 289, 11681–11694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Fichera, A.; Little, N.; Dougherty, U.; Mustafi, R.; Cerda, S.; Li, Y.C.; Delgado, J.; Arora, A.; Campbell, L.K.; Joseph, L.; et al. A Vitamin D analoguei colonic carcinogenesis in the AOM/DSS model. J. Surg. Res. 2007, 142, 239–245. [Google Scholar] [CrossRef] [PubMed]
  29. Liu, Y.; Chen, L.; Zhi, C.; Shen, M.; Sun, W.; Miao, D.; Yuan, X. 1,25(OH)2D3 deficiency induces colon inflammation via secretion of senescence-associated inflammatory cytokines. PLoS ONE 2016, 11, e0146426. [Google Scholar] [CrossRef] [Green Version]
  30. Laverny, G.; Penna, G.; Vetrano, S.; Correale, C.; Nebuloni, M.; Danese, S.; Adorini, L. Efficacy of a potent and safe vitamin D receptor agonist for the treatment of inflammatory bowel disease. Immunol. Lett. 2010, 131, 49–58. [Google Scholar] [CrossRef]
  31. Li, B.; Baylink, D.J.; Walter, M.H.; Lau, K.-H.W.; Meng, X.; Wang, J.; Cherkas, A.; Tang, X.; Qin, X. Targeted 25-hydroxyvitamin D3 1α-hydroxylase adoptive gene therapy ameliorates dss-induced colitis without causing hypercalcemia in mice. Mol. Ther. 2015, 23, 339–351. [Google Scholar] [CrossRef] [Green Version]
  32. Chen, X.-Q.; Mao, J.-Y.; Li, W.-B.; Li, J.; Yang, H.; Qian, J.-M.; Li, J.-N. Association between CYP24A1 polymorphisms and the risk of colonic polyps and colon cancer in a Chinese population. World J. Gastroenterol. 2017, 23, 5179–5186. [Google Scholar] [CrossRef]
  33. Stio, M.; Martinesi, M.; Bruni, S.; Treves, C.; D’Albasio, G.; Bagnoli, S.; Bonanomi, A.G. Interaction among vitamin D3 analogue KH 1060, TNF-α, and vitamin D receptor protein in peripheral blood mononuclear cells of inflammatory bowel disease patients. Int. Immunopharmacol. 2006, 6, 1083–1092. [Google Scholar] [CrossRef]
  34. Zhao, H.; Zhang, H.; Wu, H.; Li, H.; Liu, L.; Guo, J.; Li, C.; Shih, D.Q.; Zhang, X. Protective role of 1,25(OH)2vitamin D3 in the mucosal injury and epithelial barrier disruption in DSS-induced acute colitis in mice. BMC Gastroenterol. 2012, 12, 57. [Google Scholar] [CrossRef] [Green Version]
  35. Strauch, U.G.; Obermeier, F.; Grunwald, N.; Dunger, N.; Rath, H.C.; Scholmerich, J.; Steinmeyer, A.; Zugel, U.; Herfarth, H.H. Calcitriol analog ZK191784 ameliorates acute and chronic dextran sodium sulfate-induced colitis by modulation of intestinal dendritic cell numbers and phenotype. World J. Gastroenterol. 2007, 13, 6529–6537. [Google Scholar] [CrossRef] [PubMed]
  36. Verlinden, L.; Leyssens, C.; Beullens, I.; Marcelis, S.; Mathieu, C.; De Clercq, P.; Verstuyf, A. The vitamin D analog TX527 ameliorates disease symptoms in a chemically induced model of inflammatory bowel disease. J. Steroid Biochem. Mol. Biol. 2013, 136, 107–111. [Google Scholar] [CrossRef] [PubMed]
  37. D’Ambrosio, D.; Cippitelli, M.; Cocciolo, M.G.; Mazzeo, D.; Lucia, P.D.; Lang, R.; Sinigaglia, F.; Panina-Bordignon, P. Inhibition of IL-12 production by 1,25-dihydroxyvitamin D3. Involvement of NF-kappaB downregulation in transcriptional repression of the p40 gene. J. Clin. Investig. 1998, 101, 252–262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Du, J.; Chen, Y.; Shi, Y.; Liu, T.; Cao, Y.; Tang, Y.; Ge, X.; Nie, H.; Zheng, C.; Li, Y.C. 1,25-Dihydroxyvitamin D protects intestinal epithelial barrier by regulating the myosin light chain kinase signaling pathway. Inflamm. Bowel Dis. 2015, 21, 2495–2506. [Google Scholar] [CrossRef]
  39. Cross, H.S.; Nittke, T.; Kallay, E. Colonic vitamin D metabolism: Implications for the pathogenesis of inflammatory bowel disease and colorectal cancer. Mol. Cell. Endocrinol. 2011, 347, 70–79. [Google Scholar] [CrossRef]
  40. Sen, A.; Stark, H. Role of cytochrome P450 polymorphisms and functions in development of ulcerative colitis. World J. Gastroenterol. 2019, 25, 2846–2862. [Google Scholar] [CrossRef]
  41. Gong, C.; Long, Z.; Yu, Y.; Zhu, L.; Tian, J.; Li, S.; Li, J.; Yu, H.; Chi, Q.; Piao, D.; et al. Dietary factors and polymorphisms in vitamin D metabolism genes: The risk and prognosis of colorectal cancer in northeast China. Sci. Rep. 2017, 7, 8827. [Google Scholar] [CrossRef] [Green Version]
  42. Horváth, H.C.; Lakatos, P.; Kósa, J.P.; Bácsi, K.; Borka, K.; Bises, G.; Nittke, T.; Hershberger, P.A.; Speer, G.; Kállay, E. The candidate oncogene CYP24A1: A potential biomarker for colorectal tumorigenesis. J. Histochem. Cytochem. 2010, 58, 277–285. [Google Scholar] [CrossRef]
  43. Dong, L.M.; Ulrich, C.M.; Hsu, L.; Duggan, D.J.; Benitez, D.S.; White, E.; Slattery, M.L.; Farin, F.M.; Makar, K.W.; Carlson, C.S.; et al. Vitamin D related genes, CYP24A1 and CYP27B1, and colon cancer risk. Cancer Epidemiol. Biomark. Prev. 2009, 18, 2540–2548. [Google Scholar] [CrossRef] [Green Version]
  44. Sun, H.; Wang, C.; Hao, M.; Sun, R.; Wang, Y.; Liu, T.; Cong, X.; Liu, Y. CYP24A1 is a potential biomarker for the progression and prognosis of human colorectal cancer. Hum. Pathol. 2016, 50, 101–108. [Google Scholar] [CrossRef]
  45. Billmeier, U.; Dieterich, W.; Neurath, M.F.; Atreya, R. Molecular mechanism of action of anti-tumor necrosis factor antibodies in inflammatory bowel diseases. World J. Gastroenterol. 2016, 22, 9300–9313. [Google Scholar] [CrossRef]
  46. Xiao, Y.-T.; Yan, W.-H.; Cao, Y.; Yan, J.-K.; Cai, W. Neutralization of IL-6 and TNF-α ameliorates intestinal permeability in DSS-induced colitis. Cytokine 2016, 83, 189–192. [Google Scholar] [CrossRef] [PubMed]
  47. Fischer, A.; Gluth, M.; Pape, U.-F.; Wiedenmann, B.; Theuring, F.; Baumgart, D.C. Adalimumab prevents barrier dysfunction and antagonizes distinct effects of TNF-α on tight junction proteins and signaling pathways in intestinal epithelial cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2013, 304, G970–G979. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Landskron, G.; De la Fuente, M.; Thuwajit, P.; Thuwajit, C.; Hermoso, M.A. Chronic inflammation and cytokines in the tumor microenvironment. J. Immunol. Res. 2014, 2014, 149185. [Google Scholar] [CrossRef] [Green Version]
  49. Barbalho, S.M.; Bechara, M.D.; De Alvares Goulart, R.; Quesada, K.; Gasparini, R.G.; De Cássio Alves de Carvalho, A.; Fiorini, A.M.R. Reflections about inflammatory bowel disease and Vitamins A and D. J. Med. Food 2016, 19, 1105–1110. [Google Scholar] [CrossRef] [PubMed]
  50. Bouwmeester, T.; Bauch, A.; Ruffner, H.; Angrand, P.-O.; Bergamini, G.; Croughton, K.; Cruciat, C.; Eberhard, D.; Gagneur, J.; Ghidelli, S.; et al. A physical and functional map of the human TNF-alpha/NF-kappa B signal transduction pathway. Nat. Cell Biol. 2004, 6, 97–105. [Google Scholar] [CrossRef] [PubMed]
  51. Greten, F.R.; Eckmann, L.; Greten, T.F.; Park, J.M.; Li, Z.-W.; Egan, L.J.; Kagnoff, M.F.; Karin, M. IKKβ links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell 2004, 118, 285–296. [Google Scholar] [CrossRef] [Green Version]
  52. Wang, P.; Qiu, W.; Dudgeon, C.; Liu, H.; Huang, C.; Zambetti, G.P.; Yu, J.; Zhang, L. PUMA is directly activated by NF- κ B and contributes to TNF-α -induced apoptosis. Cell Death Differ. 2009, 16, 1192–1202. [Google Scholar] [CrossRef]
  53. Zheng, L.; Zhang, Y.-L.; Dai, Y.-C.; Chen, X.; Chen, D.-L.; Dai, Y.-T.; Tang, Z.-P. Jianpi Qingchang decoction alleviates ulcerative colitis by inhibiting nuclear factor-kB activation. World J. Gastroenterol. 2017, 23, 1180–1188. [Google Scholar] [CrossRef]
  54. Bakke, D.; Sun, J. Ancient nuclear receptor VDR with new functions: Microbiome and inflammation. Inflamm. Bowel Dis. 2018, 24, 1149–1154. [Google Scholar] [CrossRef] [Green Version]
  55. Atreya, I.; Atreya, R.; Neurath, M.F. NF-kappaB in inflammatory bowel disease. J. Intern. Med. 2008, 263, 591–596. [Google Scholar] [CrossRef] [PubMed]
  56. Wada, T.; Penninger, J.M. Mitogen-activated protein kinases in apoptosis regulation. Oncogene 2004, 23, 2838–2849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Cuenda, A.; Rousseau, S. p38 MAP-Kinases pathway regulation, function and role in human diseases. Biochim. Biophys. Acta BBA) Mol. Cell Res. 2007, 1773, 1358–1375. [Google Scholar] [CrossRef] [Green Version]
  58. Gupta, J.; Del Barco Barrantes, I.; Igea, A.; Sakellariou, S.; Pateras, I.S.; Gorgoulis, V.G.; Nebreda, A.R. Dual function of p38α MAPK in colon cancer: Suppression of colitis-associated tumor initiation but requirement for cancer cell survival. Cancer Cell 2014, 25, 484–500. [Google Scholar] [CrossRef] [Green Version]
  59. Gupta, J.; Igea, A.; Papaioannou, M.; Lopez-Casas, P.P.; Llonch, E.; Hidalgo, M.; Gorgoulis, V.G.; Nebreda, A.R. Pharmacological inhibition of p38 MAPK reduces tumor growth in patient-derived xenografts from colon tumors. Oncotarget 2015, 6, 8539–8551. [Google Scholar] [CrossRef] [Green Version]
  60. Knackstedt, R.; Shaoli, S.; Moseley, V.; Wargovich, M. The importance of the retinoid X receptor alpha in modulating inflammatory signaling in acute murine colitis. Dig. Dis. Sci. 2014, 59, 753–759. [Google Scholar] [CrossRef] [Green Version]
  61. Wu, Y.; Deng, J.; Rychahou, P.G.; Qiu, S.; Evers, B.M.; Zhou, B.P. Stabilization of snail by NF-kappaB is required for inflammation-induced cell migration and invasion. Cancer Cell 2009, 15, 416–428. [Google Scholar] [CrossRef] [Green Version]
  62. Quack, M.; Carlberg, C. The impact of functional vitamin D(3) receptor conformations on DNA-dependent vitamin D(3) signaling. Mol. Pharmacol. 2000, 57, 375–384. [Google Scholar]
  63. Long, M.D.; Sucheston-Campbell, L.E.; Campbell, M.J. Vitamin D receptor and RXR in the post-genomic era. J. Cell. Physiol. 2015, 230, 758–766. [Google Scholar] [CrossRef] [Green Version]
  64. Cohen-Lahav, M.; Shany, S.; Tobvin, D.; Chaimovitz, C.; Douvdevani, A. Vitamin D decreases NFkappaB activity by increasing IkappaBalpha levels. Nephrol. Dial. Transplant. 2006, 21, 889–897. [Google Scholar] [CrossRef] [Green Version]
  65. Kong, J.; Zhang, Z.; Musch, M.W.; Ning, G.; Sun, J.; Hart, J.; Bissonnette, M.; Li, Y.C. Novel role of the vitamin D receptor in maintaining the integrity of the intestinal mucosal barrier. Am. J. Physiol. Gastrointest. Liver Physiol. 2008, 294, G208–G216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Rubio, C.A.; Schmidt, P.T. Morphological classification of corrupted colonic crypts in ulcerative colitis. Anticancer Res. 2018, 38, 2253–2259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Hong, M.Y.; Tseng, Y.-T.; Kalaba, M.; Beidler, J. Effects of watermelon powder supplementation on colitis in high-fat diet-fed and dextran sodium sulfate-treated rats. J. Funct. Foods 2019, 54, 520–528. [Google Scholar] [CrossRef]
  68. Sun, X.; Kaufman, P.D. Ki-67: More than a proliferation marker. Chromosoma 2018, 127, 175–186. [Google Scholar] [CrossRef]
  69. Xin, Y.; He, L.; Luan, Z.; Lv, H.; Yang, H.; Zhou, Y.; Zhao, X.; Zhou, W.; Yu, S.; Tan, B.; et al. E-cadherin Mediates the Preventive Effect of Vitamin D3 in Colitis-associated Carcinogenesis. Inflamm. Bowel Dis. 2017, 23, 1535–1543. [Google Scholar] [CrossRef] [Green Version]
  70. Andersen, S.N.; Rognum, T.O.; Bakka, A.; Clausen, O.P. Ki-67: A useful marker for the evaluation of dysplasia in ulcerative colitis. Mol. Pathol. 1998, 51, 327–332. [Google Scholar] [CrossRef] [Green Version]
  71. Juniku-Shkololli, A.; Manxhuka-Kerliu, S.; Ahmetaj, H.; Khare, V.; Zekaj, S. Expression of immunohistochemical markers of progression in pre-cancerous and cancerous human colon: Correlation with serum vitamin D levels. Anticancer Res. 2015, 35, 1513–1520. [Google Scholar]
  72. Fedirko, V.; Bostick, R.M.; Flanders, W.D.; Long, Q.; Sidelnikov, E.; Shaukat, A.; Daniel, C.R.; Rutherford, R.E.; Woodard, J.J. Effects of vitamin D and calcium on proliferation and differentiation In mormal colon mucosa: A randomized clinical trial. Cancer Epidemiol. Biomark. Prev. 2009, 18, 2933–2941. [Google Scholar] [CrossRef] [Green Version]
  73. Zhu, T.; Liu, T.-J.; Shi, Y.-Y.; Zhao, Q. Vitamin D/VDR signaling pathway ameliorates 2,4,6-trinitrobenzene sulfonic acid-induced colitis by inhibiting intestinal epithelial apoptosis. Int. J. Mol. Med. 2015, 35, 1213–1218. [Google Scholar] [CrossRef] [Green Version]
  74. Zbar, A.P.; Simopoulos, C.; Karayiannakis, A.J. Cadherins: An integral role in inflammatory bowel disease and mucosal restitution. J. Gastroenterol. 2004, 39, 413–421. [Google Scholar] [CrossRef]
  75. Bostick, R.M. Effects of supplemental vitamin D and calcium on normal colon tissue and circulating biomarkers of risk for colorectal neoplasms. J. Steroid Biochem. Mol. Biol. 2015, 148, 86–95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Schneider, M.R.; Dahlhoff, M.; Horst, D.; Hirschi, B.; Trülzsch, K.; Müller-Höcker, J.; Vogelmann, R.; Allgäuer, M.; Gerhard, M.; Steininger, S.; et al. A key role for E-cadherin in intestinal homeostasis and Paneth cell maturation. PLoS ONE 2010, 5, e14325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Ooi, J.H.; Li, Y.; Rogers, C.J.; Cantorna, M.T. Vitamin D Regulates the gut microbiome and protects mice from dextran sodium sulfate–induced colitis. J. Nutr. 2013, 143, 1679–1686. [Google Scholar] [CrossRef] [PubMed]
  78. Luan, Z.; Ma, Y.; Xin, Y.; Qian, J.; Wang, H. Possible molecular mechanisms by which Vitamin D prevents inflammatory bowel disease and colitis-associated colorectal cancer. Curr. Med. Chem. 2017, 24, 911–917. [Google Scholar] [CrossRef] [PubMed]
  79. Gregorieff, A.; Pinto, D.; Begthel, H.; Destrée, O.; Kielman, M.; Clevers, H. Expression pattern of Wnt signaling components in the adult intestine. Gastroenterology 2005, 129, 626–638. [Google Scholar] [CrossRef] [PubMed]
  80. Koch, S. Extrinsic control of Wnt signaling in the intestine. Differentiation 2017, 97, 1–8. [Google Scholar] [CrossRef]
  81. Claessen, M.M.H.; Schipper, M.E.I.; Oldenburg, B.; Siersema, P.D.; Offerhaus, G.J.A.; Vleggaar, F.P. WNT-pathway activation in IBD-associated colorectal carcinogenesis: Potential biomarkers for colonic surveillance. Cell Oncol. 2010, 32, 303–310. [Google Scholar] [CrossRef]
  82. Shenoy, A.K.; Fisher, R.C.; Butterworth, E.A.; Pi, L.; Chang, L.-J.; Appelman, H.D.; Chang, M.; Scott, E.W.; Huang, E.H. Transition from colitis to cancer: High wnt activity sustains the tumor-initiating potential of colon cancer stem cell precursors. Cancer Res. 2012, 72, 5091–5100. [Google Scholar] [CrossRef] [Green Version]
  83. Keerthivasan, S.; Aghajani, K.; Dose, M.; Molinero, L.; Khan, M.W.; Venkateswaran, V.; Weber, C.; Emmanuel, A.O.; Sun, T.; Bentrem, D.J.; et al. β-Catenin promotes colitis and colon cancer through imprinting of proinflammatory properties in T cells. Sci. Transl. Med. 2014, 6, 225ra28. [Google Scholar] [CrossRef] [Green Version]
  84. Aguilera, O.; Peña, C.; García, J.M.; Larriba, M.J.; Ordóñez-Morán, P.; Navarro, D.; Barbáchano, A.; López de Silanes, I.; Ballestar, E.; Fraga, M.F.; et al. The Wnt antagonist DICKKOPF-1 gene is induced by 1α,25-dihydroxyvitamin D 3 associated to the differentiation of human colon cancer cells. Carcinogenesis 2007, 28, 1877–1884. [Google Scholar] [CrossRef]
  85. Christakos, S.; Dhawan, P.; Verstuyf, A.; Verlinden, L.; Carmeliet, G. Vitamin D: Metabolism, molecular mechanism of action, and pleiotropic effects. Physiol. Rev. 2016, 96, 365–408. [Google Scholar] [CrossRef] [PubMed]
  86. Warren, C.A.; Paulhill, K.J.; Davidson, L.A.; Lupton, J.R.; Taddeo, S.S.; Hong, M.Y.; Carroll, R.J.; Chapkin, R.S.; Turner, N.D. Quercetin may suppress rat aberrant crypt foci formation by suppressing inflammatory mediators that influence proliferation and apoptosis. J. Nutr. 2009, 139, 101–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. West, N.J.; Courtney, E.D.J.; Poullis, A.P.; Leicester, R.J. Apoptosis in the colonic crypt, colorectal adenoma, and manipulation by chemoprevention. Cancer Epidemiol. Biomark. Prev. 2009, 18, 1680–1687. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Hong, M.Y.; Turner, N.D.; Murphy, M.E.; Carroll, R.; Chapkin, R.S.; Lupton, J.R. In vivo regulation of colonic cell proliferation, differentiation, apoptosis and P27Kip1 by dietary fish oil and butyrate in rats. Cancer Prev. Res. 2015, 8, 1076–1083. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Hong, M.Y.; Lupton, J.R.; Morris, J.S.; Wang, N.; Carroll, R.J.; Davidson, L.A.; Elder, R.H.; Chapkin, R.S. Dietary fish oil reduces O6-methylguanine DNA adduct levels in rat colon in part by increasing apoptosis during tumor initiation. Cancer Epidemiol. Biomark. Prev. 2000, 9, 819–826. [Google Scholar]
  90. Yuan, B.; Zhou, S.; Lu, Y.; Liu, J.; Jin, X.; Wan, H.; Wang, F. Changes in the expression and distribution of claudins, increased epithelial apoptosis, and a mannan-binding lectin-associated immune response lead to barrier dysfunction in dextran sodium sulfate-induced rat colitis. Gut Liver 2015, 9, 734–740. [Google Scholar] [CrossRef] [Green Version]
  91. He, L.; Liu, T.; Shi, Y.; Tian, F.; Hu, H.; Deb, D.K.; Chen, Y.; Bissonnette, M.; Li, Y.C. Gut epithelial vitamin D receptor regulates microbiota-dependent mucosal inflammation by suppressing intestinal epithelial cell apoptosis. Endocrinology 2018, 159, 967–979. [Google Scholar] [CrossRef] [Green Version]
  92. Qiu, W.; Wu, B.; Wang, X.; Buchanan, M.E.; Regueiro, M.D.; Hartman, D.J.; Schoen, R.E.; Yu, J.; Zhang, L. PUMA-mediated intestinal epithelial apoptosis contributes to ulcerative colitis in humans and mice. J. Clin. Investig. 2011, 121, 1722–1732. [Google Scholar] [CrossRef]
  93. Dirisina, R.; Katzman, R.B.; Goretsky, T.; Managlia, E.; Mittal, N.; Williams, D.B.; Qiu, W.; Yu, J.; Chandel, N.S.; Zhang, L.; et al. p53 and PUMA independently regulate apoptosis of intestinal epithelial cells in patients and mice with colitis. Gastroenterology 2011, 141, 1036–1045. [Google Scholar] [CrossRef] [Green Version]
  94. Yan, F.; Polk, D.B. Probiotics: Progress toward novel therapies for intestinal diseases. Curr. Opin. Gastroenterol. 2010, 26, 95–101. [Google Scholar] [CrossRef] [Green Version]
  95. Yan, F.; Cao, H.; Cover, T.L.; Washington, M.K.; Shi, Y.; Liu, L.; Chaturvedi, R.; Peek, R.M.; Wilson, K.T.; Polk, D.B. Colon-specific delivery of a probiotic-derived soluble protein ameliorates intestinal inflammation in mice through an EGFR-dependent mechanism. J. Clin. Investig. 2011, 121, 2242–2253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Wu, S.; Yoon, S.; Zhang, Y.-G.; Lu, R.; Xia, Y.; Wan, J.; Petrof, E.O.; Claud, E.C.; Chen, D.; Sun, J. Vitamin D receptor pathway is required for probiotic protection in colitis. Am. J. Physiol. Gastrointest. Liver Physiol. 2015, 309, G341–G349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Strober, W.; Fuss, I.; Mannon, P. The fundamental basis of inflammatory bowel disease. J. Clin. Investig. 2007, 117, 514–521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Garg, M.; Hendy, P.; Ding, J.N.; Shaw, S.; Hold, G.; Hart, A. The effect of Vitamin D on intestinal inflammation and faecal microbiota in patients with ulcerative colitis. J. Crohns Colitis 2018, 12, 963–972. [Google Scholar] [CrossRef] [PubMed]
  99. Bashir, M.; Prietl, B.; Tauschmann, M.; Mautner, S.I.; Kump, P.K.; Treiber, G.; Wurm, P.; Gorkiewicz, G.; Högenauer, C.; Pieber, T.R. Effects of high doses of vitamin D3 on mucosa-associated gut microbiome vary between regions of the human gastrointestinal tract. Eur. J. Nutr. 2016, 55, 1479–1489. [Google Scholar] [CrossRef] [Green Version]
  100. Shang, M.; Sun, J. Vitamin D/VDR, probiotics, and gastrointestinal diseases. Curr. Med. Chem. 2017, 24, 876–887. [Google Scholar] [CrossRef]
  101. Issa, M.; Vijayapal, A.; Graham, M.B.; Beaulieu, D.B.; Otterson, M.F.; Lundeen, S.; Skaros, S.; Weber, L.R.; Komorowski, R.A.; Knox, J.F.; et al. Impact of clostridium difficile on inflammatory bowel disease. Clin. Gastroenterol. Hepatol. 2007, 5, 345–351. [Google Scholar] [CrossRef]
  102. Ananthakrishnan, A.N.; Cagan, A.; Gainer, V.S.; Cheng, S.-C.; Cai, T.; Szolovits, P.; Shaw, S.Y.; Churchill, S.; Karlson, E.W.; Murphy, S.N.; et al. Higher plasma vitamin D is associated with reduced risk of Clostridium difficile infection in patients with inflammatory bowel diseases. Aliment. Pharmacol. Ther. 2014, 39, 1136–1142. [Google Scholar] [CrossRef] [Green Version]
  103. Abt, M.C.; McKenney, P.T.; Pamer, E.G. Clostridium difficile colitis: Pathogenesis and host defence. Nat. Rev. Microbiol. 2016, 14, 609–620. [Google Scholar] [CrossRef]
  104. Tran, D.H.-N.; Wang, J.; Ha, C.; Ho, W.; Mattai, S.A.; Oikonomopoulos, A.; Weiss, G.; Lacey, P.; Cheng, M.; Shieh, C.; et al. Circulating cathelicidin levels correlate with mucosal disease activity in ulcerative colitis, risk of intestinal stricture in Crohn’s disease, and clinical prognosis in inflammatory bowel disease. BMC Gastroenterol. 2017, 17, 63. [Google Scholar] [CrossRef]
  105. Gubatan, J.; Mehigan, G.A.; Villegas, F.; Mitsuhashi, S.; Longhi, M.S.; Malvar, G.; Csizmadia, E.; Robson, S.; Moss, A.C. Cathelicidin mediates a protective role of vitamin D in ulcerative colitis and human colonic epithelial cells. Inflamm. Bowel Dis. 2020, 18, izz330. [Google Scholar] [CrossRef] [PubMed]
  106. Wu, S.; Zhang, Y.-G.; Lu, R.; Xia, Y.; Zhou, D.; Petrof, E.O.; Claud, E.C.; Chen, D.; Chang, E.B.; Carmeliet, G.; et al. Intestinal epithelial vitamin D receptor deletion leads to defective autophagy in colitis. Gut 2015, 64, 1082–1094. [Google Scholar] [CrossRef]
  107. Kumari, R.; Ahuja, V.; Paul, J. Fluctuations in butyrate-producing bacteria in ulcerative colitis patients of North India. World J. Gastroenterol. 2013, 19, 3404–3414. [Google Scholar] [CrossRef] [PubMed]
  108. Lagishetty, V.; Misharin, A.V.; Liu, N.Q.; Lisse, T.S.; Chun, R.F.; Ouyang, Y.; McLachlan, S.M.; Adams, J.S.; Hewison, M. Vitamin D deficiency in mice impairs colonic antibacterial activity and predisposes to colitis. Endocrinology 2010, 151, 2423–2432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Frank, D.N.; Amand, A.L.S.; Feldman, R.A.; Boedeker, E.C.; Harpaz, N.; Pace, N.R. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc. Natl. Acad. Sci. USA 2007, 104, 13780–13785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Bamola, V.D.; Ghosh, A.; Kapardar, R.K.; Lal, B.; Cheema, S.; Sarma, P.; Chaudhry, R. Gut microbial diversity in health and disease: Experience of healthy Indian subjects, and colon carcinoma and inflammatory bowel disease patients. Microb. Ecol. Health Dis. 2017, 28, 1322447. [Google Scholar] [CrossRef] [Green Version]
  111. Schaffler, H.; Herlemann, D.P.; Klinitzke, P.; Berlin, P.; Kreikemeyer, B.; Jaster, R.; Lamprecht, G. Vitamin D administration leads to a shift of the intestinal bacterial composition in Crohn’s disease patients, but not in healthy control. J. Dig. Dis. 2018, 19, 225–234. [Google Scholar] [CrossRef]
  112. Nielsen, O.H.; Rejnmark, L.; Moss, A.C. Role of vitamin D in the natural history of inflammatory bowel disease. J. Crohns Colitis 2018, 12, 742–752. [Google Scholar] [CrossRef]
  113. Lips, P. Relative value of 25(OH)D and 1,25(OH)2D measurements. J. Bone Miner. Res. 2007, 22, 1668–1671. [Google Scholar] [CrossRef]
  114. Sahota, O. Understanding vitamin D deficiency. Age Ageing 2014, 43, 589–591. [Google Scholar] [CrossRef] [Green Version]
Table 1. Characteristics of Published Clinical Trials Examining Vitamin D and Inflammation.
Table 1. Characteristics of Published Clinical Trials Examining Vitamin D and Inflammation.
Author (Year)Study Type, PopulationMeasurementsResultsConclusion
Brozek et al. (2012) [24]Factor and cluster analysis of 105 patients undergoing primary curative surgery for adenocarcinomasExpression of VDR, CYP27B1, CYP24A1 and COX-2 in relation to tumor grade, anatomical location, and genderCompared to adjacent mucosa mRNA expression in cancerous lesions was ↑ in CYP27B1 (4-fold in low/high grade cancers), CYP24A1 (20-35-fold in low/high grade lesions) and COX-2 (2-fold in high grade cancers) but not VDR; in distal colon tumors CYP27B1 expression 2-fold ↑ in men than womenAntagonism between COX-2 and Vitamin D could be important factor in epithelial colorectal cancer cell growth; differences in COX-2 expression could influence variation in incidence at different anatomical subsites; cancer incidence gender-specific differences correlate with age
Frăţilă & Iliaş (2013) [25]Retrospective study
2006–2010
80 patients w/ longstanding ulcerative colitis (LUC) dx (42 women, 38 men)
53.5 ± 14.2 years old
Colonoscopy biopsies: 20 high-grade dysplasia (HGD), 20 low grade dysplasia (LGD), 20 regenerative atypia, 20 indefinite for dysplasia;
immunohistochemical (IHC) staining applied for Ki-67, clone MIB-1, and anti-COX-2
COX-2 is positive in 72.5% LUC biopsies; LGD and HGD had Ki-67 stainingIHC staining may be used to manage increased colorectal cancer risk in LUC patients
Leedham et al. (2009) [26]Mutation burden analysis of individual crypts across colitis associated neoplasmsPCR and sequencing analysis to establish individual crypt adenomatous polyposis coli (APC), p53, K-RAS, and 17p loss of heterozygosity mutation burdenMonoclonality observed in most lesions typically from p53 lesion and occasionally K-RASp53 mutation was the initiator in the majority of lesions; K-RAS activation found to be gatekeeping mutation
Wang et al. (2014) [27]Macrophages from COX-2 KO and COXNeo/Neo miceCOX-2 expression and PG expression in the presence and absence of LPS stimulation1,25-OHD results in dose-dependent inhibition of COX-2 expression and phosphorylation of Akt and IκBα in murine macrophages with and without LPS stimulationVitamin D influences inflammation and supplementation could improve chronic inflammatory diseases via targeting THEM4/Akt/NF-κB signaling
Hummel et al. (2014) [16]Adenocarcinoma cell line COGA-1A culture and treatmentCOGA-1A cells treated with 10 nM 1,25-OHD, 100 ng/mL IL-6, 50 ng/mL or combination of for 6, 12, and 24 hrs; total RNA isolation; reverse transcription polymerase chain reaction (RT-PCR)COGA-1A cells + 1,25-OHD = ↑ VDR expression; IL-6 ↑ CYP24A1 expression 3x; COX-2 and 15-PGH expression “unresponsive” with 1,25-OHD in COGA-1A cells, but TNF-α “highly ↑ COX-2 expression”TNF-α and IL-6 inhibited vit D expression-activating gene CYP27B1 in COGA-1A cells
Fichera et al. (2007) [28]Male A/J mice (25 g)
AOM/DSS-induced colitis
Vitamin D analog Ro26-2198 (0.01 µg/kg body wt/day x 28 days); severity of colitis assessed via the Disease Activity Index; hematoxylin and eosin colonic sections examined for dysplasia; colonic lysates assessed for c-Myc, COX-2, phosphor-(active) extracellular signal regulated kinase (ERK) via Western blottingDSS treatment ↑ c-Myc 15-fold, ERK 10-fold, COX-2 2.5-fold
Ro26-2198 ↓ proliferative (c-Myc, ERK) and pro-inflammatory (COX-2) signals and dysplasia progression
Vitamin D analog can be considered when treating colitis
Liu et al. (2016) [29]25-OHD 1α-hydroxylase knockout (Cyp27b1−/−) mice fed high calcium, phosphate, and lactose rescue dietBody weight, colon length, and colonic histologic structureCyp27b1-/- had ↓ bodyweight, colon length, colon length to bodyweight ratio, mucosa thickness and ↑ crypt damage1,25-OHD may influence colon inflammation and cancer development and progression
Laverny et al. (2010) [30]Peripheral blood mononuclear cells (PBMC) obtained from IBD patients (21 ulcerative colitis patients, 22 Crohn’s Disease patients)
26 males, 13 females;
C57BL/6 mice w/ 3% DSS treatment; cell study
VDR agonist BXL-62 administration; histology; cytokine quantification; RT-PCRBXL-62 has 3x less calcemic activity, ↓ proinflammatory cytokines in cells of IBD patients, ↑ CYP24A1 expression, ameliorates experimental colitis compared to 1,25-OHDBXL-62 is a VDR agonist that does not promote hypercalcemia, ↓ in vitro pro-inflammatory cytokines, and may be used as IBD treatment
Li et al. (2015) [31]Gene therapy (regulate CYP27B1 expression via CD11b+/Gr1+ monocytes) in murine DSS-induced IBD modelSurvival rate, weight, colonic structure, mucosal regeneration index, cytokine expression↑ survival, body weight gain, colon length, mucosal regeneration and ↓ proinflammatory cytokines (TNF-α, IL-1β, IL-6, IL-12, IL-23, Th1 and Th17)This preliminary evidence of a monocyte-based adoptive CYP27B1 gene therapy in a murine IBD model may lead to novel therapy for autoimmune disease like IBD
Chen et al. (2017) [32] Genotyping of patients with colonic polyps (n = 144), colon cancer (n = 96), and UC (n = 44) to determine correlations between CYP24A1 SNPs and diseasesGenotyping of four SNPs (rs4809957, rs6068816, rs6091822 and rs8124792) and their association with colonic polyps, colon cancer, and UCCYP24A1 polymorphisms rs4809957 A/G and rs6068816 C/T were associated with colonic polyps, colon cancer, and UCrs4809957, rs6068816, rs6091822 and rs8124792 are related to risk of colonic polyps and cancer; the A allele in rs8124792 may indicate colonic polyps and cancer not UC
Stio et al. (2006) [33]PBMC of IBD patients; KH 1060 (vitamin D analog) treatment[3H]thymidine incorporation; ELISA kit; VDR levels w/ Western blottingKH 1060 ↓ PBMC proliferation, ↓ TNF-α, ↑ VDR expressionKH 1060 can be used on IBD patients to ↓ TNF-α
Zhao et al. (2012) [34]C57BL/6 mice (n = 30), equally divided among 3 groups: 2% DSS treatment, control, and vitamin DSerum vitamin D levels, Rachmilewitz DAI (disease activity), colonic injury/inflammation; myeloperoxidase (MPO) activity, mesenteric lymph nodes cells (MLNs) and LPMC isolation; immunohistochemistry; immunofluorescence; in vivo/in vitro permeability; TEER measurement; qT-PCR; Western blottingSerum vitamin D levels were ↓ in DSS group compared to control; DSS damaged mucosal barrier and ↑ colonic inflammation, ↑ weight loss; vitamin D inhibited colonic inflammation, maintained intestinal permeability, preserved colonic structural integrity, prevented DSS-induced tight junction disruptionVitamin D warrants therapeutic potential against IBD
Strauch et al. (2007) [35]Female Balb/c mice w/ 3% DSS treatment; ZK191784 (vitamin D analog) 100 μg/kg per dayHistological examination; ELISA of MLNs; RT-PCR; immunohistochemistryZK191784 treatment ameliorates acute and chronic DSS-induced colitis, suppresses proinflammatory cytokine secretion by MLNs and primary dendritic cellsAnother study by which vitamin D analog is shown to be beneficial against experimental colitis
Verlinden et al. (2013) [36]DSS-induced colitis in mice; TX527 (vitamin D analog)Histological examination, transcript levels of proinflammatory cytokinesTX527 ameliorated DSS-colitis symptoms by ↓ diarrhea and bleeding, ↓ mucosal damage, crypt loss, immune cell infiltration, and ↓ proinflammatory cytokines (IL-1, IL-6, IFN-γ and TNF-α)TX527 suggests therapeutic potential for IBD management
D’Ambrosio et al. (1998) [37]Cotransfected monocytic RAW264.7 cells with p40 promotor/reporter constructs and expression vectors of VDR and RXR-αExpression of NF-κB pathway and cytokines following 1,25-OHD1,25-OH D caused downregulation of NF-κB activation and ↓ in downstream IL-121,25-OHD may downregulate IL-12 production via downregulation of NF-kB activation and p40-kB sequence binding
Du et al. (2015) [38]Cell study (HCT116, Caco-2, SW480) w/ TNF-α ± 1,25-OHD treatment;
C57BL/6 mice w/ paricalcitol (vitamin D analog) treatment;
Human colonic mucosal biopsies
Histology/immunostaining; Western blotting, RT-PCR, chromatin immunoprecipitation assay, myeloid perioxidase assayTNF-α ↓ cells’ transepithelial electrical resistance (TER); > 70% VDR ↓ unable to counteract TNF-α activity and maintain TER; 1,25-OHD counteracts NF-κB by suppressing the myosin light chain kinase signaling pathway (MLCK); MLCK pathway activation = ↓ VDR in human biopsies; paricalcitol inhibits MLCK activation, regulates mucosal barrier, and ameliorates colitis in the mouse modelVDR is required to maintain and protect intestinal epithelial barrier
Meeker et al. (2014) [8]Smad3−/− mice fed w/ maintenance diet, high vitamin D diet (5 IU/g), or vitamin D-deficient (1 IU/g) diet infected with ~2 × 107 CFU Hb in Brucella broth or Brucella broth alone (control)Helicobacter bilis infection; Fecal polymerase chain reaction; serum vitamin D and calcium; histopathology; immunochemistry; epithelial and lamina propria leukocyte cell samples; Western blotting↑ vitamin D diet after H. bilis infection = ↑ serum vitamin D w/o affecting calcium levels. ↓ dysplasia, ↓ inflammation (MAPK, NF-κB), ↓ IBD scoresVitamin D intake suggests reduced colitis symptoms and delay progression to early stage carcinogenesis
Knackstedt et al. (2013) [13]Female C57BL/6J mice fed normal chow or vitamin D-deficient diet;
Acute colitis: 10 mg/kg AOM + 2% DSS treatment
Colitis-associated cancer (CAC): 12 mg/kg AOM + 4% DSS treatment
Vitamin D quantification; colitis scoring; immunohistochemistry; protein extraction; immunoblotting; RT-PCRAOM/DSS-induced acute colitis mice had ↓ body weight, ↑ blood loss, shortened colon, ↓ serum vitamin D compared to control; CAC model had mortality rate between 10–40%, shortened colon, ↓ serum vitamin D, TNF-α upregulated, RXRα and VDR downregulatedVitamin D-deficient diet leads to increased colitis scores in the mouse model, downregulated RXRα, VDR, and decreased serum vitamin D levels

Share and Cite

MDPI and ACS Style

Garcia, P.M.; Moore, J.; Kahan, D.; Hong, M.Y. Effects of Vitamin D Supplementation on Inflammation, Colonic Cell Kinetics, and Microbiota in Colitis: A Review. Molecules 2020, 25, 2300. https://doi.org/10.3390/molecules25102300

AMA Style

Garcia PM, Moore J, Kahan D, Hong MY. Effects of Vitamin D Supplementation on Inflammation, Colonic Cell Kinetics, and Microbiota in Colitis: A Review. Molecules. 2020; 25(10):2300. https://doi.org/10.3390/molecules25102300

Chicago/Turabian Style

Garcia, Patricia Mae, Jeff Moore, David Kahan, and Mee Young Hong. 2020. "Effects of Vitamin D Supplementation on Inflammation, Colonic Cell Kinetics, and Microbiota in Colitis: A Review" Molecules 25, no. 10: 2300. https://doi.org/10.3390/molecules25102300

Article Metrics

Back to TopTop