Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Anticancer Research
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Anticancer Research

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Visit us on Facebook
  • Follow us on Linkedin
Research ArticleProceedings of the Joint International Symposium “Vitamin D in Prevention and Therapy” and “Biologic Effects of Light”, 5-7 June, 2019 (Homburg/Saar, Germany)

The Effect of Various Doses of Oral Vitamin D3 Supplementation on Gut Microbiota in Healthy Adults: A Randomized, Double-blinded, Dose-response Study

NIPITH CHAROENNGAM, ARASH SHIRVANI, TYLER A. KALAJIAN, ANJELI SONG and MICHAEL F. HOLICK
Anticancer Research January 2020, 40 (1) 551-556; DOI: https://doi.org/10.21873/anticanres.13984
NIPITH CHAROENNGAM
Department of Medicine, Section of Endocrinology, Nutrition, and Diabetes, Vitamin D, Skin and Bone Research Laboratory, Boston University Medical Center, Boston, MA, U.S.A.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
ARASH SHIRVANI
Department of Medicine, Section of Endocrinology, Nutrition, and Diabetes, Vitamin D, Skin and Bone Research Laboratory, Boston University Medical Center, Boston, MA, U.S.A.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
TYLER A. KALAJIAN
Department of Medicine, Section of Endocrinology, Nutrition, and Diabetes, Vitamin D, Skin and Bone Research Laboratory, Boston University Medical Center, Boston, MA, U.S.A.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
ANJELI SONG
Department of Medicine, Section of Endocrinology, Nutrition, and Diabetes, Vitamin D, Skin and Bone Research Laboratory, Boston University Medical Center, Boston, MA, U.S.A.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
MICHAEL F. HOLICK
Department of Medicine, Section of Endocrinology, Nutrition, and Diabetes, Vitamin D, Skin and Bone Research Laboratory, Boston University Medical Center, Boston, MA, U.S.A.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: mfholick{at}bu.edu
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background/Aim: To investigate the effects of vitamin D3 supplementation on gut microbiota. Patients and Methods: Twenty adults with vitamin D insufficiency/deficiency [25(OH)D <30 ng/ml] were enrolled and given 600, 4,000 or 10,000 IUs/day of oral vitamin D3. Stool samples were collected at baseline and 8 weeks for identifying gut microbiota using 16S rRNA gene amplification and sequencing. Results: Baseline serum 25(OH)D was associated with increased relative abundance of Akkermansia and decreased relative abundance of Porphyromonas (p<0.05). After the intervention, we observed a dose-dependent increase in relative abundance of Bacteroides with a significant difference between the 600 IUs and the 10,000 IUs groups (p=0.027), and Parabacteroides with a significant difference between the 600 IUs and the 4,000 IUs groups (p=0.039). Conclusion: Increased serum 25(OH)D was associated with increased beneficial bacteria and decreased pathogenic bacteria. A dose-dependent increase in bacteria associated with decreased inflammatory bowel disease activity was observed after vitamin D3 supplementation.

  • Vitamin D3
  • 25-hydroxyivitamin D
  • Vitamin D3 supplementation
  • gut microbiota
  • randomized clinical trial

Vitamin D plays an essential role in regulating calcium and phosphate metabolism and maintaining healthy mineralized skeleton (1). Observational studies have suggested that high serum 25-hydroxyvitamin D [25(OH)D] is associated with a variety of extra-skeletal outcomes, including increased longevity and prevention of cardiovascular diseases, metabolic syndrome, cancers, and autoimmune diseases (1-3). However, whether or not the causality can be drawn from this association is largely controversial, and through which mechanisms does vitamin D exert its extra-skeletal effects is still unclarified.

It has been suggested that alteration in gut microbiota is one of the mechanisms that link vitamin D with some of its extra-skeletal clinical outcomes. Variation in gut microbiota has been linked with a number of physiological functions, including absorption and metabolism of nutrients, immune regulation, and biosynthesis of neurotransmitters and hormones (4). The probable mechanism by which vitamin D may affect gut microbiota is mainly mediated by the biologic effect of 1,25-dihydroxyvitamin D [1,25(OH)2D], the active form of vitamin D, on host immune response (4, 5). 1,25(OH)2D induces macrophages to produce antimicrobial peptides, resulting in bacterial killing. On the other hand, 1,25(OH)2D also modulates cell-mediated immune response by inhibiting proliferation of T cells, inducing a shift from Th1 to a Th2 development, suppressing interferon-γ and interleukin-17 producing T cells, facilitating T regulatory cells, thereby decreasing inflammatory activity against certain bacteria (4, 5).

Evidence on the effect of vitamin D supplementation on human gut microbiome is relatively limited. There are a few clinical trials including randomized and non-randomized interventional studies reporting inconsistent results, although most of them reported significant changes in certain bacteria (6-10). Therefore, as of now, the effect on vitamin D on human gut microbiota is still undetermined and inconclusive. The aim of this study was to investigate the effects of various doses of oral vitamin D3 supplementation on the composition of gut microbiota in healthy adults. This will provide some perspective on how vitamin D could possibly exert extra-skeletal effects through alterations of gut microbiota.

Patients and Methods

Healthy adults who had vitamin D insufficiency/deficiency [25(OH)D<30 ng/ml] were enrolled in a randomized, double-blinded, investigator initiated, pilot study. This study investigated broad gene expression changes in peripheral blood mononuclear cells following vitamin D3 supplementation of 600, 4,000 or 10,000 IU/day of vitamin D3, as previously described (11). The study was approved by the Boston University Medical Campus Institutional Review Board (NCT01696409, H-35506).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Relative abundance of Akkermansia spp. (left) and Porphyromonas spp. (right) of the participants at various levels of serum 25(OH)D at baseline of the study.

Stool microbiota assessment. Stool samples were collected at baseline of the study and at 8 weeks after vitamin D supplementation using Explorer™ Microbiome Sampling Kit. Samples were subsequently sent for microbiome analysis using 16S rRNA gene amplification and sequencing at uBiome Inc., (San Francisco, CA, USA). Data are reported as inverse Simpson diversity index and the relative abundance of a set of microbial taxa known to be associated with health conditions, as previously described (12).

Statistical analysis. Data were expressed as mean±standard deviation (SD). Pearson correlation analysis was used for determining correlation between two parametric continuous variables. Differences in relative abundance of each bacteria before and after the intervention were analyzed using paired T-test. Comparisons in changes in relative abundance of bacteria among the different arms were analyzed using one-way analysis of variance (ANOVA), followed by post hoc least significant difference (LSD) test to estimate the pairwise significance of difference. All statistical analyses were performed using a Statistical Package for the Social Sciences (SPSS) version 25.

Results

Among the 33 participants who were enrolled in the study, a total of 20 participants had fecal microbiome data for analysis (600 IU/day, N=7; 4,000 IU/day, N=7; 10,000 IU/day, N=6). After 8 weeks of the study intervention, participants receiving 600 IUs/day raised their serum 25(OH)D levels from 16.9±6.0 ng/ml to 20.0±3.4 ng/ml although without statistical significance (p=0.153). Participants receiving 4,000 IUs/day raised their serum 25(OH)D levels from 20.3±6.3 ng/ml to 39.0±8.7 ng/ml (p=0.01). Participants receiving 10,000 IUs/day raised their serum 25(OH)D levels from 18.5±3.5 ng/ml to 67.3±3.1 ng/ml (p<0.001).

As shown in Figure 1, we observed that baseline serum 25(OH)D levels were significantly positively correlated with relative abundance of Akkermansia spp. (R=0.684, p=0.001), and negatively correlated with relative abundance of Porphyromonas spp. (R=-0.435, p=0.043). We also observed a significant positive correlation between serum 25(OH)D and phylum Verrucomicrobia (R=0.680, p=0.001). In addition, there is a significant positive correlation between serum parathyroid hormone and relative abundance of Bacteroides spp. (R=0.535, p=0.012, Figure 1B), Campylobacter spp. (R=0.564, p=0.008), Family Enterobacteriaceae (R=0.468, p=0.032), class Gammaproteobacteria (R=0.538, p=0.012), and Bacteroidia (R=0.484, p=0.026), phylum Bacteroidetes (R=0.490, p=0.024), and Proteobacteria (R=0.584, p=0.005). There is a significant negative correlation between serum parathyroid hormone and relative abundance of class Negativicutes (R=−0.510, p=0.018) and Clostridia (R=−0.513, p=0.017), and phylum Firmicutes (R=−0.545, p=0.011).

Figure 2 demonstrates the relative abundance of each bacterial genus before and after 8 weeks of intervention in all participants regardless of arm. After 8 weeks of intervention, we found a statistically significant decrease in the relative abundance of Faecalibacterium spp., class Clostridia and family Ruminococcaceae (p<0.05 all). Moreover, we observed a significant decrease in Firmicutes to Bacteroidetes (F/B) ratio (2.9±1.6 to 1.9±1.0, p=0.024). Alpha-diversity did not significantly change in any group after the 8-week intervention.

Figure 3 demonstrates the comparisons of changes in relative abundance of each bacterial genus among the three arms of different doses of vitamin D3. We observed a dose-dependent increase in relative abundance of Bacteroides spp. with a statistically significant difference between the 600 IUs group and the 10,000 IUs group (p=0.027). Moreover, a similar dose-dependent increase in relative abundance of Parabacteroides spp. was observed with a statistically significant difference between the 600 IUs group and the 4,000 IUs group (p=0.039).

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Relative abundance of clinically relevant bacterial genera before and after 8 weeks of intervention in all participants. Data are expressed as mean±SEM.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Changes in relative abundance of clinically relevant bacterial genera in participants receiving 600 IUs/day (left), 4,000 IUs/day (middle), and 10,000 IUs/day (right) of vitamin D3 for 8 weeks. Data are expressed as mean±SEM.

Discussion

Vitamin D is known to modulate innate and adaptive immune response through several mechanisms. Monocytes express the enzyme 25-hydroxyvitamin D-1α-hydroxylase (CYP27B1) that converts 25(OH)D into the active form of 1,25(OH)2D, which induces macrophages to produce antimicrobial peptides, resulting in bacterial killing (5). 1,25(OH)2D also modulates T cell functions by inhibiting T cell proliferation, inducing a shift from Th1 to a Th2 development, suppressing Th17 cell development and facilitating regulatory T cells (5). Therefore, it is likely that vitamin D, by modulating host immune response against certain bacteria, may affect the composition of gut microbiota. To investigate this, we conducted a randomized, double-blinded, dose-response, pilot study aiming to determine the effects of various doses of oral vitamin D3 (600 IUs/day, 4,000 IUs/day, and 10,000 IUs/day) on gut microbiota in healthy adults. Correlation analysis at baseline of the study showed that the relative abundance of a variety of bacteria are associated with serum 25(OH)D and PTH levels. After the 8-week intervention, we observed a statistically significant decrease in Firmicutes to Bacteroidetes ratio in all subjects and significant dose-dependent increase in relative abundance of Bacteroides spp. and Parabacteroides spp. These results provided some insight into how vitamin D status is possibly related to several clinical outcomes via its interaction with gut microbiota.

We observed that baseline serum 25(OH)D levels were positively correlated with relative abundance of Akkermansia spp. (R=0.684, p=0.001). This result is consistent with an observation by Su et al. that vitamin D receptor knockout mice as well as vitamin D deficient mice receiving high fat diet had diminished gut Akkermansia municiphila (13). Increased richness of Akkermansia municiphila has been shown to be associated with decreased risk of cancer, obesity and atherosclerosis, as well as improved fasting plasma glucose, plasma triglycerides and body fat mobilization (14-16). This is possibly mediated by its interaction with the intestinal epithelium causing improvement of gut barrier function and decreased metabolic endotoxemia (14-16). The increased relative abundance of this bacteria in association with higher serum 25(OH)D level, therefore, could be another explanation for how vitamin D is related to cardio-metabolic disorders and cancers. After vitamin D treatment for 8 weeks, regardless of dosage, a significant decrease in F/B ratio was observed. High F/B ratio has been linked to obesity and poor glycemic control (17). This may be another mechanism explaining the possible relationship between vitamin D deficiency and obesity, insulin resistance, and metabolic syndrome (18).

We found an inverse correlation between 25(OH)D levels and relative abundance of Porphyromonas spp., which is supportive of the finding by Grenier et al. (19) that 1,25(OH)2D could directly inhibit growth and expression of the virulence factor of Porphyromonas gingivalis, a causative organism of chronic periodontitis. This finding could not only explain the link between vitamin D deficiency and periodontitis reported in several observational studies (20), but also supports the result from a randomized clinical trial by Hiremath et al. which has demonstrated the possible benefit of vitamin D in treating gingivitis (21).

We observed a substantial dose-dependent increase in the relative abundance of Bacteroides spp. and Parabacteroides spp. after 8 weeks of vitamin D supplementation. It has been shown that Bacteroides and Parabacteroides were suppressed in patients with active inflammatory bowel diseases compared with normal controls (22, 23). Suppression of these two bacteria is believed to play a role in the pathogenesis of inflammatory bowel disease. The likely mechanism is that these bacteria help maintain the expansion of regulatory T cells, which, in turn, alleviate intestinal inflammation (24-26). This observation could therefore indicate another mechanism explaining the findings from clinical trials demonstrating an efficacy of vitamin D treatment in controlling the relapse rate in patients with inflammatory bowel diseases (27). In fact, the association between vitamin D status and Bacteroides and Parabacteroides is still inconclusive, since animal studies have reported inconsistent results (4, 28-31). One clinical trial did report an increase in the relative abundance of Parabacteroides in patients with Crohn's disease but not in healthy individuals during the third and fourth weeks of high dose oral vitamin D3 supplementation (20,000 IUs per day for 3 days then every other day for 4 weeks) (10). Further studies are warranted to evaluate the association of vitamin D status with gut microbiota, and inflammatory bowel disease.

It should be acknowledged that our study has some limitations. Since this is a pilot study with a relatively small sample size, further studies with larger sample size should be conducted to confirm our findings. Furthermore, it might require a longer period of time to demonstrate a clinically significant and more robust change in the composition of gut microbiota in response to oral vitamin D supplementation. A study that evaluates both short-term and long-term effects of vitamin D on gut microbiota is required.

Conclusion

In conclusion, we observed that an increase in baseline serum 25(OH)D levels was correlated with increased bacteria associated with decreased risk of cardiovascular and metabolic diseases, obesity, and cancers. We also found that increased baseline 25(OH)D levels were inversely correlated with decreased periodondopathic bacteria. After 8 weeks of vitamin D supplementation, we observed an alteration of gut microbiota towards a decrease in Firmicutes to Bacteroidetes ratio, which is an indicator associated with obesity and metabolic syndrome. Finally, we observed a dose-dependent increase in bacteria associated with decreased inflammatory bowel disease activity in response to various doses of vitamin D3 supplementation.

Footnotes

  • Authors' Contributions

    Conceptualization: N. Charoenngam, A. Shirvani, T.K. Kalajian, M.F. Holick; Collecting data: T. Kalajian, A. Song; Data analysis: N. Charoenngam, A. Shirvani, A. Song; Writing - original draft: N. Charoenngam, A. Shirvani; Visualization: N. Charoenngam, A. Shirvani; Writing - review & editing: N. Charoenngam, A. Shirvani, M.F. Holick.

  • Conflicts of Interest

    Michael F. Holick is a consultant for Quest Diagnostics Inc. and Ontometrics Inc, and on the speaker's Bureau for Abbott Inc. The remaining Authors declare no conflict of interest regarding this study.

  • Received November 21, 2019.
  • Revision received November 27, 2019.
  • Accepted December 2, 2019.
  • Copyright© 2020, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved

References

  1. ↵
    1. Holick MF
    : Vitamin d deficiency. N Engl J Med 357(3): 266-281, 2007. PMID: 17634462. DOI: 10.1056/NEJMra070553
    OpenUrlCrossRefPubMed
    1. Dudenkov DV,
    2. Mara KC,
    3. Petterson TM,
    4. Maxson JA,
    5. Thacher TD
    : Serum 25-hydroxyvitamin d values and risk of all-cause and cause-specific mortality: A population-based cohort study. Mayo Clin Proc 93(6): 721-730, 2018. PMID: 29730089. DOI: 10.1016/j.mayocp.2018.03.006
    OpenUrlPubMed
  2. ↵
    1. Rosen CJ,
    2. Adams JS,
    3. Bikle DD,
    4. Black DM,
    5. Demay MB,
    6. Manson JE,
    7. Murad MH,
    8. Kovacs CS
    : The nonskeletal effects of vitamin d: An endocrine society scientific statement. Endocr Rev 33(3): 456-492, 2012. PMID: 22596255. DOI: 10.1210/er.2012-1000
    OpenUrlCrossRefPubMed
  3. ↵
    1. Waterhouse M,
    2. Hope B,
    3. Krause L,
    4. Morrison M,
    5. Protani MM,
    6. Zakrzewski M,
    7. Neale RE
    : Vitamin d and the gut microbiome: A systematic review of in vivo studies. Eur J Nutr 58(7): 2895-2910, 2019. PMID: 30324342. DOI: 10.1007/s00394-018-1842-7
    OpenUrlPubMed
  4. ↵
    1. Aranow C
    : Vitamin d and the immune system. J Investig Med 59(6): 881-886, 2011. PMID: 21527855. DOI: 10.2310/JIM.0b013e31821b8755
    OpenUrlAbstract/FREE Full Text
  5. ↵
    1. Bashir M,
    2. Prietl B,
    3. Tauschmann M,
    4. Mautner SI,
    5. Kump PK,
    6. Treiber G,
    7. Wurm P,
    8. Gorkiewicz G,
    9. Hogenauer C,
    10. Pieber TR
    : Effects of high doses of vitamin d3 on mucosa-associated gut microbiome vary between regions of the human gastrointestinal tract. Eur J Nutr 55(4): 1479-1489, 2016. PMID: 26130323. DOI: 10.1007/s00394-015-0966-2
    OpenUrlCrossRefPubMed
    1. Cantarel BL,
    2. Waubant E,
    3. Chehoud C,
    4. Kuczynski J,
    5. DeSantis TZ,
    6. Warrington J,
    7. Venkatesan A,
    8. Fraser CM,
    9. Mowry EM
    : Gut microbiota in multiple sclerosis: Possible influence of immunomodulators. J Investig Med 63(5): 729-734, 2015. PMID: 25775034. DOI: 10.1097/jim.0000000000000192
    OpenUrlAbstract/FREE Full Text
    1. Garg M,
    2. Hendy P,
    3. Ding JN,
    4. Shaw S,
    5. Hold G,
    6. Hart A
    : The effect of vitamin d on intestinal inflammation and faecal microbiota in patients with ulcerative colitis. J Crohns Colitis 12(8): 963-972, 2018. PMID: 29726893. DOI: 10.1093/ecco-jcc/jjy052
    OpenUrlPubMed
    1. Kanhere M,
    2. He J,
    3. Chassaing B,
    4. Ziegler TR,
    5. Alvarez JA,
    6. Ivie EA,
    7. Hao L,
    8. Hanfelt J,
    9. Gewirtz AT,
    10. Tangpricha V
    : Bolus weekly vitamin d3 supplementation impacts gut and airway microbiota in adults with cystic fibrosis: A double-blind, randomized, placebo-controlled clinical trial. J Clin Endocrinol Metab 103(2): 564-574, 2018. PMID: 29726893. DOI: 10.1210/jc.2017-01983
    OpenUrlCrossRefPubMed
  6. ↵
    1. Schaffler H,
    2. Herlemann DP,
    3. Klinitzke P,
    4. Berlin P,
    5. Kreikemeyer B,
    6. Jaster R,
    7. Lamprecht G
    : Vitamin d administration leads to a shift of the intestinal bacterial composition in crohn's disease patients, but not in healthy controls. J Dig Dis 19(4): 225-234, 2018. PMID: 29573237., DOI: 10.1111/1751-2980.12591
    OpenUrlCrossRefPubMed
  7. ↵
    1. Shirvani A,
    2. Kalajian TA,
    3. Song A,
    4. Holick MF
    : Disassociation of vitamin d's calcemic activity and non-calcemic genomic activity and individual responsiveness: A randomized controlled double-blind clinical trial. Sci Rep 9(1): 17685, 2019. DOI: 10.1038/s41598-019-53864-1
    OpenUrlCrossRefPubMed
  8. ↵
    1. Vera-Wolf P,
    2. Cárdenas JP,
    3. Morton AM,
    4. Norambuena T,
    5. Torres R,
    6. Leon LE,
    7. Bik EM,
    8. Ugalde JA,
    9. Almonacid DE,
    10. Richman J,
    11. Apte ZS
    : Measures of reproducibility in sampling and laboratory processing methods in high-throughput microbiome analysis. bioRxiv: 322677, 2018. DOI: 10.1101/322677
  9. ↵
    1. Su D,
    2. Nie Y,
    3. Zhu A,
    4. Chen Z,
    5. Wu P,
    6. Zhang L,
    7. Luo M,
    8. Sun Q,
    9. Cai L,
    10. Lai Y,
    11. Xiao Z,
    12. Duan Z,
    13. Zheng S,
    14. Wu G,
    15. Hu R,
    16. Tsukamoto H,
    17. Lugea A,
    18. Liu Z,
    19. Pandol SJ,
    20. Han YP
    : Vitamin d signaling through induction of paneth cell defensins maintains gut microbiota and improves metabolic disorders and hepatic steatosis in animal models. Front Physiol 7: 498, 2016. PMID: 27895587. DOI: 10.3389/fphys.2016.00498
    OpenUrlCrossRefPubMed
  10. ↵
    1. Everard A,
    2. Belzer C,
    3. Geurts L,
    4. Ouwerkerk JP,
    5. Druart C,
    6. Bindels LB,
    7. Guiot Y,
    8. Derrien M,
    9. Muccioli GG,
    10. Delzenne NM,
    11. de Vos WM,
    12. Cani PD
    : Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci USA 110(22): 9066-9071, 2013. PMID: 23671105. DOI: 10.1073/pnas.1219451110
    OpenUrlAbstract/FREE Full Text
    1. Ma J,
    2. Li H
    : The role of gut microbiota in atherosclerosis and hypertension. Front Pharmacol 9: 1082, 2018. PMID: 30319417. DOI: 10.3389/fphar.2018.01082
    OpenUrlCrossRefPubMed
  11. ↵
    1. Naito Y,
    2. Uchiyama K,
    3. Takagi T
    : A next-generation beneficial microbe: Akkermansia muciniphila. J Clin Biochem Nutr 63(1): 33-35, 2018. PMID: 30087541. DOI: 10.3164/jcbn.18-57
    OpenUrlCrossRefPubMed
  12. ↵
    1. Castaner O,
    2. Goday A,
    3. Park Y-M,
    4. Lee S-H,
    5. Magkos F,
    6. Shiow S-ATE,
    7. Schröder H
    : The gut microbiome profile in obesity: A systematic review. Int J Endocrinol 2018: 4095789-4095789, 2018. PMID: 29849617. DOI: 10.1155/2018/4095789
    OpenUrlCrossRefPubMed
  13. ↵
    1. Wieder-Huszla S,
    2. Jurczak A,
    3. Szkup M,
    4. Barczak K,
    5. Dołęgowska B,
    6. Schneider-Matyka D,
    7. Owsianowska J,
    8. Grochans E
    : Relationships between vitamin d3 and metabolic syndrome. Int J Environ Res Public Health 16(2): 175, 2019. PMID: 30634516. DOI: 10.3390/ijerph16020175
    OpenUrlPubMed
  14. ↵
    1. Grenier D,
    2. Morin MP,
    3. Fournier-Larente J,
    4. Chen H
    : Vitamin d inhibits the growth of and virulence factor gene expression by Porphyromonas gingivalis and blocks activation of the nuclear factor kappa b transcription factor in monocytes. J Periodontal Res 51(3): 359-365, 2016. PMID: 26297053. DOI: 10.1111/jre.12315
    OpenUrlCrossRefPubMed
  15. ↵
    1. Jagelaviciene E,
    2. Vaitkeviciene I,
    3. Silingaite D,
    4. Sinkunaite E,
    5. Daugelaite G
    : The relationship between vitamin d and periodontal pathology. Medicina (Kaunas) 54(3), 2018. PMID: 30344276. DOI: 10.3390/medicina54030045
  16. ↵
    1. Hiremath VP,
    2. Rao CB,
    3. Naiak V,
    4. Prasad KV
    : Anti-inflammatory effect of vitamin d on gingivitis: A dose response randomised controlled trial. Indian J Public Health 57(1): 29-32, 2013. PMID: 23507683. DOI: 10.4103/0019-557x.111365
    OpenUrlPubMed
  17. ↵
    1. Olbjorn C,
    2. Cvancarova Smastuen M,
    3. Thiis-Evensen E,
    4. Nakstad B,
    5. Vatn MH,
    6. Jahnsen J,
    7. Ricanek P,
    8. Vatn S,
    9. Moen AEF,
    10. Tannaes TM,
    11. Lindstrom JC,
    12. Soderholm JD,
    13. Halfvarson J,
    14. Gomollon F,
    15. Casen C,
    16. Karlsson MK,
    17. Kalla R,
    18. Adams AT,
    19. Satsangi J,
    20. Perminow G
    : Fecal microbiota profiles in treatment-naive pediatric inflammatory bowel disease - associations with disease phenotype, treatment, and outcome. Clin Exp Gastroenterol 12: 37-49, 2019. PMID: 30774408. DOI: 10.2147/ceg.s186235
    OpenUrlCrossRefPubMed
  18. ↵
    1. Zhou Y,
    2. Zhi F
    : Lower level of bacteroides in the gut microbiota is associated with inflammatory bowel disease: A meta-analysis. Biomed Res Int 2016: 5828959, 2016. PMID: 27999802. DOI: 10.1155/2016/5828959
    OpenUrlPubMed
  19. ↵
    1. Round JL,
    2. Mazmanian SK
    : Inducible foxp3+ regulatory t-cell development by a commensal bacterium of the intestinal microbiota. Proc Natl Acad Sci USA 107(27): 12204-12209, 2010. PMID: 20566854. DOI: 10.1073/pnas.0909122107
    OpenUrlAbstract/FREE Full Text
    1. Kverka M,
    2. Zakostelska Z,
    3. Klimesova K,
    4. Sokol D,
    5. Hudcovic T,
    6. Hrncir T,
    7. Rossmann P,
    8. Mrazek J,
    9. Kopecny J,
    10. Verdu EF,
    11. Tlaskalova-Hogenova H
    : Oral administration of parabacteroides distasonis antigens attenuates experimental murine colitis through modulation of immunity and microbiota composition. Clin Exp Immunol 163(2): 250-259, 2011. PMID: 21087444. DOI: 10.1111/j.1365-2249.2010.04286.x
    OpenUrlCrossRefPubMed
  20. ↵
    1. Mayne CG,
    2. Williams CB
    : Induced and natural regulatory t cells in the development of inflammatory bowel disease. Inflam Bowel Dis 19(8): 1772-1788, 2013. PMID: 23656897. DOI: 10.1097/MIB.0b013e318281f5a3
    OpenUrlCrossRefPubMed
  21. ↵
    1. Li J,
    2. Chen N,
    3. Wang D,
    4. Zhang J,
    5. Gong X
    : Efficacy of vitamin d in treatment of inflammatory bowel disease: A meta-analysis. Medicine (Baltimore) 97(46): e12662, 2018. PMID: 30431562. DOI: 10.1097/md.0000000000012662
    OpenUrlPubMed
  22. ↵
    1. Wu S,
    2. Zhang YG,
    3. Lu R,
    4. Xia Y,
    5. Zhou D,
    6. Petrof EO,
    7. Claud EC,
    8. Chen D,
    9. Chang EB,
    10. Carmeliet G,
    11. Sun J
    : Intestinal epithelial vitamin d receptor deletion leads to defective autophagy in colitis. Gut 64(7): 1082-1094, 2015. PMID: 25080448. DOI: 10.1136/gutjnl-2014-307436
    OpenUrlAbstract/FREE Full Text
    1. Jahani R,
    2. Fielding KA,
    3. Chen J,
    4. Villa CR,
    5. Castelli LM,
    6. Ward WE,
    7. Comelli EM
    : Low vitamin d status throughout life results in an inflammatory prone status but does not alter bone mineral or strength in healthy 3-month-old cd-1 male mice. Mol Nutr Food Res 58(7): 1491-1501, 2014. PMID: 24823836. DOI: 10.1002/mnfr.201300928
    OpenUrlPubMed
    1. Villa CR,
    2. Taibi A,
    3. Chen J,
    4. Ward WE,
    5. Comelli EM
    : Colonic bacteroides are positively associated with trabecular bone structure and programmed by maternal vitamin d in male but not female offspring in an obesogenic environment. Int J Obes (Lond) 42(4): 696-703, 2018. PMID: 29188819. DOI: 10.1038/ijo.2017.294
    OpenUrlPubMed
  23. ↵
    1. Jin D,
    2. Wu S,
    3. Zhang YG,
    4. Lu R,
    5. Xia Y,
    6. Dong H,
    7. Sun J
    : Lack of vitamin d receptor causes dysbiosis and changes the functions of the murine intestinal microbiome. Clin Ther 37(5): 996-1009.e1007, 2015. PMID: 26046242. DOI: 10.1016/j.clinthera.2015.04.004
    OpenUrlPubMed
View Abstract
PreviousNext
Back to top

In this issue

Anticancer Research
Vol. 40, Issue 1
January 2020
  • Table of Contents
  • Table of Contents (PDF)
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Anticancer Research.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
The Effect of Various Doses of Oral Vitamin D3 Supplementation on Gut Microbiota in Healthy Adults: A Randomized, Double-blinded, Dose-response Study
(Your Name) has sent you a message from Anticancer Research
(Your Name) thought you would like to see the Anticancer Research web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
17 + 1 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
The Effect of Various Doses of Oral Vitamin D3 Supplementation on Gut Microbiota in Healthy Adults: A Randomized, Double-blinded, Dose-response Study
NIPITH CHAROENNGAM, ARASH SHIRVANI, TYLER A. KALAJIAN, ANJELI SONG, MICHAEL F. HOLICK
Anticancer Research Jan 2020, 40 (1) 551-556; DOI: 10.21873/anticanres.13984

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
The Effect of Various Doses of Oral Vitamin D3 Supplementation on Gut Microbiota in Healthy Adults: A Randomized, Double-blinded, Dose-response Study
NIPITH CHAROENNGAM, ARASH SHIRVANI, TYLER A. KALAJIAN, ANJELI SONG, MICHAEL F. HOLICK
Anticancer Research Jan 2020, 40 (1) 551-556; DOI: 10.21873/anticanres.13984
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Patients and Methods
    • Results
    • Discussion
    • Conclusion
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • The Relationship Between Oxidative Stress, Selenium, and Cumulative Risk in Metabolic Syndrome
  • Google Scholar

More in this TOC Section

  • Improving Photoprotection and Implications for 25(OH)D Formation
  • Relevance of Vitamin D in Melanoma Development, Progression and Therapy
  • Sunbeds and Melanoma Risk: Many Open Questions, Not Yet Time to Close the Debate
Show more Proceedings of the Joint International Symposium “Vitamin D in Prevention and Therapy” and “Biologic Effects of Light”, 5-7 June, 2019 (Homburg/Saar, Germany)

Similar Articles

Keywords

  • Vitamin D3
  • 25-hydroxyivitamin D
  • Vitamin D3 supplementation
  • gut microbiota
  • randomized clinical trial
Anticancer Research

© 2025 Anticancer Research

Powered by HighWire