Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2922 2023-08-07 12:55:04 |
2 format change + 4 word(s) 2926 2023-08-08 03:10:09 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Muñoz-Garach, A.; García-Fontana, B.; Muñoz-Torres, M. Vitamin D and Type 2 Diabetes. Encyclopedia. Available online: https://encyclopedia.pub/entry/47738 (accessed on 09 May 2024).
Muñoz-Garach A, García-Fontana B, Muñoz-Torres M. Vitamin D and Type 2 Diabetes. Encyclopedia. Available at: https://encyclopedia.pub/entry/47738. Accessed May 09, 2024.
Muñoz-Garach, Araceli, Beatriz García-Fontana, Manuel Muñoz-Torres. "Vitamin D and Type 2 Diabetes" Encyclopedia, https://encyclopedia.pub/entry/47738 (accessed May 09, 2024).
Muñoz-Garach, A., García-Fontana, B., & Muñoz-Torres, M. (2023, August 07). Vitamin D and Type 2 Diabetes. In Encyclopedia. https://encyclopedia.pub/entry/47738
Muñoz-Garach, Araceli, et al. "Vitamin D and Type 2 Diabetes." Encyclopedia. Web. 07 August, 2023.
Vitamin D and Type 2 Diabetes
Edit

The management and early treatment of type 2 diabetes (T2D) are essential to prevent further complications involving loss of quality of life and premature death. It is unclear whether vitamin D deficiency might be contributing to an increased T2D risk. A vast body of evidence associates vitamin D deficiency and T2D. This relationship could be mediated by the direct and indirect effects of vitamin D on glucose homeostasis such as insulin secretion, insulin sensitivity, and systemic inflammation.

dairy products vitamin D type 2 diabetes

1. Vitamin D Physiology and Glucose Homeostasis

The term vitamin D includes vitamin D2 or ergocalciferol and vitamin D3 or cholecalciferol. The main metabolites of vitamin D, which differ in their hydroxylation patterns, are 25-hydroxyvitamin D or calcidiol (25(OH)D) and 1,25-dihydroxyvitamin D3 or calcitriol (1,25-(OH)2D3). In humans, the main sources of vitamin D come from the skin through the cutaneous synthesis of vitamin D3 and, to a lesser extent, from the intake of foods rich in vitamins D2 and D3 or supplements. Circulating vitamin D is bound to vitamin D binding protein (DBP), which transports it to the liver, there vitamin D25-hydroxylase converts it to 25(OH)D. This form of vitamin D is primarily converted to the most biologically active form, 1,25-(OH)2D in the kidneys. This transformation is done by the enzyme 25-hydroxyvitamin D-1alpha-hydroxylase (CYP27B1). The presence of CYP27B1 in multiple tissues, which also express the vitamin D receptor, suggests that vitamin D could play an important function beyond bone metabolism.
Both in vitro and in vivo studies have reported that vitamin D may play an important role in the maintenance of pancreatic beta cell function [1]. This effect could have different explanations. It could be induced by the activation of the vitamin D receptor (VDR) located in pancreatic beta cells. It was suggested by the study results that showed how mice without VDR have impaired insulin secretion [2] and the addition of calcitriol to the culture medium stimulated pancreatic islets and resulted in an increased insulin secretion [3].
Moreover, vitamin D could also influence insulin secretion by regulating calcium channel opening and closure. Calcitriol participates as a chemical messenger interacting with different receptors regulating calcium flux in beta cells. They are located on the phospholipid layers of plasma membranes. For this reason, calcium is essential for appropriate insulin secretion by pancreatic beta cells; and, therefore, insufficient vitamin D may alter normal insulin secretion through alterations in calcium flux in beta cells [4][5]. In relation to this, the regulation of the protein calbindin, a calcium-binding protein, by vitamin D may be another mechanism influencing insulin secretion. In addition, preclinical studies show that vitamin D can reduce the hyperactivity of the renin angiotensin system and, thus, improve the functioning of beta cells (Leung PS. Nutrients 2016 [6]).
An adequate vitamin D level can also improve insulin resistance pathways associated with diabetes. It is caused mainly by alterations in calcium flux and concentration through the cell membranes of insulin-responsive tissues [7]. The regulation of extracellular and intracellular calcium concentrations may promote dephosphorylation of glucose transporter-4 (GLUT-4) driving a reduced insulin-stimulated glucose transport [5][8]. The 1,25-(OH)2D stimulates the expression of insulin receptors and, therefore, stimulates insulin sensitivity. In addition, calcitriol could also improve insulin sensitivity activating the peroxisome proliferator-activated delta receptor (PPAR-d), a transcription factor that regulates fatty acids metabolism in adipose tissue and the skeletal muscle. Another interesting study indicates that insulin resistance may also be reduced by the specific effects of calcitriol on hepatic lipid synthesis and glucose output, and on skeletal muscle (Leung PS. Nutrients 2016 [6]).
Calcitriol has a central role in a wide variety of metabolic pathways by binding to the VDR, and the measurement of its substrate 25(OH)D is an important marker for health risks. This receptor is expressed in an assortment of cells, such as in the pancreatic beta cells of Langerhans, but also in liver, adipose tissue, and muscle cells [9][10]. The VDR and the 1α-hydroxylase, the enzyme catalyzing calcidiol to calcitriol conversion, are expressed in primary preadipocytes and recently differentiated adipocytes [9]. Therefore, in vitro studies suggest that calcitriol regulates the growth of human adipose tissue and its remodeling. Moreover, fat tissue is a storage site for vitamin D [10]. In contrast, a higher body mass index (BMI) is associated to lower vitamin D concentrations. Vitamin D, a fat-soluble hormone, is sequestered in the adipose tissue and, consequently, only small quantities are available for circulation [11]. On the other hand, since the concentrations of 25(OH)D in serum and adipose tissue are closely related, obesity can reduce serum 25(OH)D through volumetric dilution and the distribution of 25(OH)D in larger fat volumes [12].
Vitamin D could also shorten the effects of chronic inflammation, and it is well established that it plays a key role in the pathogenesis of T2D. Therefore, 1,25(OH)2D can protect against cytokine-induced apoptosis of beta cells directly regulating the activity and expression of cytokines, with an improvement in insulin sensitivity [13]. Moreover, vitamin D demonstrated the possibility of deactivating inflammatory cytokines associated with insulin resistance and promoting calbindin expression which involves protection from apoptosis [14]. Finally, vitamin D also reduces the accumulation of advanced glycation products in experimental studies [15]. These products are related with the development of T2D complications and have been involved with insulin resistance. Vitamin D functions related with glucose homeostasis are summarized in Figure 1.
Figure 1. Vitamin D functions related with glucose homeostasis.

2. Vitamin D Status and Its Relationship with T2D in Cross-Sectional and Longitudinal Studies

Serum 25(OH)D concentrations have been noticed to be inversely associated with glucose homeostasis, insulin resistance, and beta cell function, and forecast lower risks of both metabolic syndrome and T2D [16][17][18]. Numerous clinical studies have associated vitamin D inadequacy with the development of insulin resistance in different populations, not only in adults [19][20][21] but also in children [22][23].
Consistently, higher baseline 25(OH)D levels have been found to predict better beta cell function and lower glucose levels in subjects at risk for T2D in longitudinal studies [24]. Overall, data from observational studies strongly support an association between low vitamin D status and incidence of T2D [25][26][27].
The researchers discuss in this entry the largest prospective articles and some meta-analyses. In 2013, Afzal et al. published the results of a prospective cohort study that included 9841 participants who were followed-up for 29 years. They found an odds ratio for the development of T2D of 1.5 (95% CI 1.33–1.70) between the lowest and the highest quartile of 25(OH)D [26]. More recently, Park et al. measured 25(OH)D levels in a cohort of 903 adults without diabetes or prediabetes, these authors found an inverse dose-response association between 25(OH)D concentration and risk of diabetes. They proposed a target 25(OH)D of 50 ng/mL; higher than the levels previously suggested in other studies, in the attempt to influence and reduce the incidence rate of diabetes [28]. These data are consistent with the levels published recently by Avila-Rubio et al. in postmenopausal women, the authors link values of 25(OH)D > 45 ng/dL in these women with better glycemic indexes measured by homeostasis model assessment (HOMA) [29].
The multicenter EPIC-InterAct study measured plasma 25(OH)D metabolites: non-epimeric 25(OH)D3, 3-epi-25(OH)D3 and 25(OH)D2. They identified that plasma non-epimeric 25(OH)D3 (the major component of total 25(OH)D) was inversely associated with T2D, whereas 3-epi-25(OH)D3 was positively associated with the incidence of T2D, and 25(OH)D2 was not associated with T2D [30].
Another large cohort was The Melbourne cohort, which included a sample of middle-aged Australians, the authors showed how vitamin D status was inversely associated with the risk of T2D and, apparently, this association cannot be explained by reverse causality [31]. If the association was due to reverse causality, then a much stronger association would be expected to be observed in the first few years of follow-up.
The meta-analysis conducted by Song et al. included 21 observational studies with 76,220 subjects in total; the authors found a 38% lesser risk of developing T2D in the highest baseline reference category of 25(OH)D compared to the lowest one (95% CI 0.54–0.70) [27].
Despite the consistency of these results, all these were observational studies and estimation of causality cannot be completely excluded because of residual confounding agents.

3. Vitamin D Supplementation and Risk of T2D: Randomized Trials and Meta-Analysis

In the last decade, more than ten well-designed, randomized trials evaluated the effect of vitamin D3 supplementation on glucose homeostasis in subjects at risk for T2D and showed inconsistent results. The researchers have selected a set of studies that analyzed outcomes related to the objectives of this reseach. Table 1 summarizes the main results of these studies.
Table 1. Clinical trials investigating the association between vitamin D supplementation and risk of type 2 diabetes (T2D).
BMI, body mass index; FPG, fasting plasma glucose; 2hs PG, 2 h plasma glucose; HbA1c, glycated hemoglobin; HOMA-IR, homeostatic model assessment of insulin resistance; IFG, impaired fasting glucose; IGT, impaired glucose tolerance; NAFLD, nonalcoholic fatty liver disease; T2D, type 2 diabetes.
In Table 1 the researchers describe the main findings of the largest trials. Sollid et al. [37] conducted a randomized clinical trial with approximately 500 prediabetes subjects comparing vitamin D versus placebo for the prevention of T2D. They supplemented with 20,000 IU cholecalciferol weekly and after one year, no significant differences were reported between those receiving vitamin D and those taking placebo in any of the glycemic or inflammatory markers and blood pressure, regardless of baseline serum 25(OH)D concentrations.
Two years later, Forouhi et al. [33] compared, in another large randomized trial including 340 prediabetes or at risk of developing T2D subjects, the effect of supplementation with cholecalciferol or ergocalciferol (both 100,000 IU/month) versus placebo, for four months. Prediabetes was estimated by the Cambridge Risk Score [45]. Despite vitamin D supplementation, neither ergocalciferol or cholecalciferol, resulted in increased 25(OH)D2 and 25(OH)D3 concentrations. No differences in HbA1c concentration were found between groups. It is important to point out that only half of the subjects had concentrations of 25(OH)D < 50 nmol/L. This data could influence the results.
Their results are in accordance with previous findings by Davidson et al. [41]. They supplemented a cohort of Latino and African Americans subjects with prediabetes and hypovitaminosis D at baseline for one year. They used a cholecalciferol dose sufficient to raise serum 25(OH)D levels into the upper-normal range versus placebo. They did not find any effect on insulin secretion or sensitivity, nor the proportion of subjects who developed T2D or whose oral glucose tolerance test became normal [41].
However, it is possible to find some positive effect on glycemic markers in some studies. In this line, Gagnon et al. [36] reported when they performed a post hoc analysis only including subjects with prediabetes, an improvement in insulin sensitivity indices was observed. Previously, they gave a supplement of calcium carbonate 1200 mg and cholecalciferol 2000–6000 UI daily to subjects with glucose intolerance or recently diagnosed diabetes, but they found no effect on insulin sensitivity or secretion, and beta cell function. So, despite most clinical randomized trials failing to show a favorable effect of vitamin D supplementation on glycemic control, insulin sensitivity indices, and incident T2D [33][34][35][36][37][38][39][41][43] in subjects at risk for diabetes, there is some interesting evidence supporting a beneficial effect of vitamin D on beta cell function. In fact, Mitri et al. [42] reported in 2011 a significant improvement in insulin secretion in 92 prediabetic subjects who were overweight or obese and at risk for T2D. They were supplemented with cholecalciferol 2000 IU daily and calcium carbonate versus placebo for four months. An important restriction of the above described studies is that they were not designed specifically to assess glycemic homeostasis and the results found correspond to post hoc analyses.
Although not being designed for this purpose, the researchers would like to point out the results of the Vitamin D and Omega-3 Trial (VITAL), a large-scale trial that evaluated high-dose vitamin D supplementation. This research was designed to evaluate the effect of supplementation with vitamin D on incidence of invasive cancer or cardiovascular events versus placebo. Overall, no differences were found between groups. However, in Black Americans a potential beneficial effect was found in cancer mortality [46].
A recent meta-analysis conducted by Rafiq et al. showed an inverse relationship; higher vitamin D concentrations were associated with lower BMI in T2D patients and non-diabetic subjects at risk for T2D. But this association was more pronounced in T2D patients. Moreover, the correlation was directly associated to the BMI quartiles, so the highest BMI quartile had the greatest correlations in both populations, both T2D and non-diabetic [47].
Tang et al. [48] published a meta-analysis and did not find an effect of vitamin D supplementation on the incidence of T2D. However, the authors suggested a possible dose-response effect of vitamin D supplementation to improve glucose and insulin metabolism among non-diabetic adults. They postulated a possible benefit of taking vitamin D supplements in higher doses for the primary prevention of T2D.
In summary, studies were very heterogeneous in terms of design, duration, and type of supplement administered and participants characteristics. It is noteworthy that adherence to the treatment would have played a major role in arguing these results.
Recently, the design of a new randomized clinical trial has been published and its results can be a determinant in clarifying many of the uncertainties that exist today. The D2d is a large randomized clinical trial (including participants from 22 sites across the U.S.) hypothesizing that supplementation with vitamin D3 daily lowers risk of diabetes in adults with prediabetes [32]. This trial meets people with a large spectrum of diabetes risk, more convenient for testing the underlying hypothesis. D2d trial results are expected to answer two important questions: whether vitamin D supplementation is useful to prevent T2D and how the 2010 expanded American Diabetes Association (ADA) criteria for prediabetes would impact the natural history of this state previous to diabetes.

4. New Thresholds for the Relationship between Vitamin D and T2D

An important question that has arisen is what 25(OH)D levels are necessary to influence glycemic homeostasis and the risk of developing T2D. Three recent studies have addressed this issue. Von Horst et al. found that optimal 25OHD concentrations for reducing IR were around 50 ng/dL in a randomized controlled study with 81 Asian women [43]. Avila Rubio et al. [29], in a study conducted in women with postmenopausal osteoporosis, suggested that the established goal of reaching a level of 25(OH)D > 30 ng/mL was insufficient to improve glucose metabolism in these population. The data from this study indicates that 25(OH)D > 45 ng/mL are necessary to achieve this goal. These data are consistent with a cohort study of 903 adults of 12 years of duration in non-diabetic population where reaching values of 25(OH)D > 50 ng/mL contributed to reach the maximum benefits to reduce the risk of incident diabetes [28]. Therefore, it is important to establish what 25(OH)D values are necessary to achieve and, even more importantly, maintain all the potential benefits of vitamin D. The currently available studies do not allow us to answer this question with certainty.

References

  1. Cade, C.; Norman, A.W. Rapid normalization/stimulation by 1,25- dihydroxyvitamin d3 of insulin secretion and glucose tolerance in the vitamin d-deficient rat. Endocrinology 1987, 120, 1490–1497.
  2. Zeitz, U.; Weber, K.; Soegiarto, D.W.; Wolf, E.; Balling, R.; Erben, R.G. Impaired insulin secretory capacity in mice lacking a functional vitamin D receptor. FASEB J. 2003, 17, 509–511.
  3. Bouillon, R.; Carmeliet, G.; Verlinden, L.; Van Etten, E.; Verstuyf, A.; Luderer, H.F.; Lieben, L.; Mathieu, C.; Demay, M. Vitamin D and human health: Lessons from vitamin D receptor null mice. Endocr. Rev. 2008, 29, 726–776.
  4. Bland, R.; Markovic, D.; Hills, C.E.; Hughes, S.V.; Chan, S.L.F.; Squires, P.E.; Hewison, M. Expression of 25-hydroxyvitamin D3-1alpha-hydroxylase in pancreatic islets. J. Steroid Biochem. Mol. Biol. 2004, 89–90, 121–125.
  5. Reusch, J.E.; Begum, N.; Sussman, K.E.; Draznin, B. Regulation of GLUT-4 phosphorylation by intracellular calcium in adipocytes. Endocrinology 1991, 129, 3269–3273.
  6. Leung, P.S. The Potential Protective Action of Vitamin D in Hepatic Insulin Resistance and Pancreatic Islet Dysfunction in Type 2 Diabetes Mellitus. Nutrients 2016, 8, 147.
  7. Wright, D.C.; Hucker, K.A.; Holloszy, J.O.; Han, D.H. Ca2+ and AMPK both mediate stimulation of glucose transport by muscle contractions. Diabetes 2004, 53, 330–335.
  8. Draznin, B. Cytosolic calcium and insulin resistance. Am. J. Kidney Dis. 1993, 21, 32–38.
  9. Nimitphong, H.; Holick, M.F.; Fried, S.K.; Lee, M.-J. 25-hydroxyvitamin D(3) and 1,25-dihydroxyvitamin D(3) promote the differentiation of human subcutaneous preadipocytes. PLoS ONE 2012, 7, e52171.
  10. Blum, M.; Dolnikowski, G.; Seyoum, E.; Harris, S.S.; Booth, S.L.; Peterson, J.; Saltzman, E.; Dawson-Hughes, B. Vitamin D(3) in fat tissue. Endocrine 2008, 33, 90–94.
  11. Wamberg, L.; Christiansen, T.; Paulsen, S.K.; Fisker, S.; Rask, P.; Rejnmark, L.; Richelsen, B.; Pedersen, S.B. Expression of vitamin D-metabolizing enzymes in human adipose tissue—The effect of obesity and diet-induced weight loss. Int. J. Obes. 2013, 37, 651–657.
  12. Hyppönen, E.; Boucher, B.J. Adiposity, vitamin D requirements, and clinical implications for obesity-related metabolic abnormalities. Nutr. Rev. 2018, 76, 678–692.
  13. Chun, R.F.; Liu, P.T.; Modlin, R.L.; Adams, J.S.; Hewison, M. Impact of vitamin D on immune function: lessons learned from genome-wide analysis. Front. Physiol. 2014, 5, 151.
  14. Christakos, S.; Liu, Y. Biological actions and mechanism of action of calbindin in the process of apoptosis. J. Steroid Biochem. Mol. Biol. 2004, 89–90, 401–404.
  15. Salum, E.; Kals, J.; Kampus, P.; Salum, T.; Zilmer, K.; Aunapuu, M.; Arend, A.; Eha, J.; Zilmer, M. Vitamin D reduces deposition of advanced glycation end-products in the aortic wall and systemic oxidative stress in diabetic rats. Diabetes Res. Clin. Pract. 2013, 100, 243–249.
  16. Forouhi, N.G.; Luan, J.; Cooper, A.; Boucher, B.J.; Wareham, N.J. Baseline serum 25-hydroxy vitamin d is predictive of future glycemic status and insulin resistance: The Medical Research Council Ely Prospective Study 1990-2000. Diabetes 2008, 57, 2619–2625.
  17. Kayaniyil, S.; Vieth, R.; Retnakaran, R.; Knight, J.A.; Qi, Y.; Gerstein, H.C.; Perkins, B.A.; Harris, S.B.; Zinman, B.; Hanley, A.J. Association of vitamin D with insulin resistance and beta-cell dysfunction in subjects at risk for type 2 diabetes. Diabetes Care 2010, 33, 1379–1381.
  18. Maki, K.C.; Fulgoni, V.L., 3rd; Keast, D.R.; Rains, T.M.; Park, K.M.; Rubin, M.R. Vitamin D intake and status are associated with lower prevalence of metabolic syndrome in U.S. adults: National Health and Nutrition Examination Surveys 2003–2006. Metab. Syndr. Relat. Disord. 2012, 10, 363–372.
  19. Chiu, K.C.; Chu, A.; Go, V.L.W.; Saad, M.F. Hypovitaminosis D is associated with insulin resistance and β cell dysfunction. Am. J. Clin. Nutr. 2004, 79, 820–825.
  20. Gannage-Yared, M.-H.; Chedid, R.; Khalife, S.; Azzi, E.; Zoghbi, F.; Halaby, G. Vitamin D in relation to metabolic risk factors, insulin sensitivity and adiponectin in a young Middle-Eastern population. Eur. J. Endocrinol. 2009, 160, 965–971.
  21. Scragg, R.; Holdaway, I.; Singh, V.; Metcalf, P.; Baker, J.; Dryson, E. Serum 25-hydroxyvitamin D3 levels decreased in impaired glucose tolerance and diabetes mellitus. Diabetes Res. Clin. Pract. 1995, 27, 181–188.
  22. Olson, M.L.; Maalouf, N.M.; Oden, J.D.; White, P.C.; Hutchison, M.R. Vitamin D deficiency in obese children and its relationship to glucose homeostasis. J. Clin. Endocrinol. Metab. 2012, 97, 279–285.
  23. Parikh, S.; Guo, D.-H.; Pollock, N.K.; Petty, K.; Bhagatwala, J.; Gutin, B.; Houk, C.; Zhu, H.; Dong, Y. Circulating 25-hydroxyvitamin D concentrations are correlated with cardiometabolic risk among American black and white adolescents living in a year-round sunny climate. Diabetes Care 2012, 35, 1133–1138.
  24. Kayaniyil, S.; Retnakaran, R.; Harris, S.B.; Vieth, R.; Knight, J.A.; Gerstein, H.C.; Perkins, B.A.; Zinman, B.; Hanley, A.J. Prospective associations of vitamin D with beta-cell function and glycemia: The PROspective Metabolism and ISlet cell Evaluation (PROMISE) cohort study. Diabetes 2011, 60, 2947–2953.
  25. Forouhi, N.G.; Ye, Z.; Rickard, A.P.; Khaw, K.T.; Luben, R.; Langenberg, C.; Wareham, N.J. Circulating 25-hydroxyvitamin D concentration and the risk of type 2 diabetes: Results from the European Prospective Investigation into Cancer (EPIC)-Norfolk cohort and updated meta-analysis of prospective studies. Diabetologia 2012, 55, 2173–2182.
  26. Afzal, S.; Bojesen, S.E.; Nordestgaard, B.G. Low 25-hydroxyvitamin D and risk of type 2 diabetes: A prospective cohort study and metaanalysis. Clin. Chem. 2013, 59, 381–391.
  27. Song, Y.; Wang, L.; Pittas, A.G.; Del Gobbo, L.C.; Zhang, C.; Manson, J.E.; Hu, F.B. Blood 25-hydroxy vitamin D levels and incident type 2 diabetes: A meta-analysis of prospective studies. Diabetes Care 2013, 36, 1422–1428.
  28. Park, S.K.; Garland, C.F.; Gorham, E.D.; BuDoff, L.; Barrett-Connor, E. Plasma 25-hydroxyvitamin D concentration and risk of type 2 diabetes and pre-diabetes: 12-year cohort study. PLoS ONE 2018, 13, e0193070.
  29. Avila-Rubio, V.; Garcia-Fontana, B.; Novo-Rodriguez, C.; Cantero-Hinojosa, J.; Reyes-Garcia, R.; Munoz-Torres, M. Higher Levels of Serum 25-Hydroxyvitamin D Are Related to Improved Glucose Homeostasis in Women with Postmenopausal Osteoporosis. J. Women’s Health 2018, 27, 1007–1015.
  30. Zheng, J.-S.; Imamura, F.; Sharp, S.J.; van der Schouw, Y.T.; Sluijs, I.; Gundersen, T.E.; Ardanaz, E.; Boeing, H.; Bonet, C.; Gómez, J.H.; et al. Association of plasma vitamin D metabolites with incident type 2 diabetes: EPIC-InterAct case-cohort study. J. Clin. Endocrinol. Metab. 2019, 104, 1293–1303.
  31. Heath, A.K.; Williamson, E.J.; Hodge, A.M.; Ebeling, P.R.; Eyles, D.W.; Kvaskoff, D.; O’Dea, K.; Giles, G.G.; English, D.R. Vitamin D status and the risk of type 2 diabetes: The Melbourne Collaborative Cohort Study. Diabetes Res. Clin. Pract. 2018, 149, 179–187.
  32. LeBlanc, E.S.; Pratley, R.E.; Dawson-Hughes, B.; Staten, M.A.; Sheehan, P.R.; Lewis, M.R.; Peters, A.; Kim, S.H.; Chatterjee, R.; Aroda, V.R.; et al. Baseline Characteristics of the Vitamin D and Type 2 Diabetes (D2d) Study: A Contemporary Prediabetes Cohort That Will Inform Diabetes Prevention Efforts. Diabetes Care 2018, 41, 1590–1599.
  33. Forouhi, N.G.; Menon, R.K.; Sharp, S.J.; Mannan, N.; Timms, P.M.; Martineau, A.R.; Rickard, A.P.; Boucher, B.J.; Chowdhury, T.A.; Griffiths, C.J.; et al. Effects of vitamin D2 or D3 supplementation on glycaemic control and cardiometabolic risk among people at risk of type 2 diabetes: Results of a randomized double-blind placebo-controlled trial. Diabetes Obes. Metab. 2016, 18, 392–400.
  34. Wagner, H.; Alvarsson, M.; Mannheimer, B.; Degerblad, M.; Ostenson, C.-G. No Effect of High-Dose Vitamin D Treatment on beta-Cell Function, Insulin Sensitivity, or Glucose Homeostasis in Subjects With Abnormal Glucose Tolerance: A Randomized Clinical Trial. Diabetes Care 2016, 39, 345–352.
  35. Tuomainen, T.-P.; Virtanen, J.K.; Voutilainen, S.; Nurmi, T.; Mursu, J.; de Mello, V.D.F.; Schwab, U.; Hakumaki, M.; Pulkki, K.; Uusitupa, M. Glucose Metabolism Effects of Vitamin D in Prediabetes: The VitDmet Randomized Placebo-Controlled Supplementation Study. J. Diabetes Res. 2015, 2015, 672653.
  36. Gagnon, C.; Daly, R.M.; Carpentier, A.; Lu, Z.X.; Shore-Lorenti, C.; Sikaris, K.; Jean, S.; Ebeling, P.R. Effects of combined calcium and vitamin D supplementation on insulin secretion, insulin sensitivity and beta-cell function in multi-ethnic vitamin D-deficient adults at risk for type 2 diabetes: A pilot randomized, placebo-controlled trial. PLoS ONE 2014, 9, e109607.
  37. Sollid, S.T.; Hutchinson, M.Y.S.; Fuskevag, O.M.; Figenschau, Y.; Joakimsen, R.M.; Schirmer, H.; Njolstad, I.; Svartberg, J.; Kamycheva, E.; Jorde, R. No effect of high-dose vitamin D supplementation on glycemic status or cardiovascular risk factors in subjects with prediabetes. Diabetes Care 2014, 37, 2123–2131.
  38. Oosterwerff, M.M.; Eekhoff, E.M.; Van Schoor, N.M.; Boeke, A.J.P.; Nanayakkara, P.; Meijnen, R.; Knol, D.L.; Kramer, M.H.; Lips, P. Effect of moderate-dose vitamin D supplementation on insulin sensitivity in vitamin D-deficient non-Western immigrants in the Netherlands: A randomized placebo-controlled trial. Am. J. Clin. Nutr. 2014, 100, 152–160.
  39. Salehpour, A.; Shidfar, F.; Hosseinpanah, F.; Vafa, M.; Razaghi, M.; Amiri, F. Does vitamin D3 supplementation improve glucose homeostasis in overweight or obese women? A double-blind, randomized, placebo-controlled clinical trial. Diabet. Med. 2013, 30, 1477–1481.
  40. Belenchia, A.M.; Tosh, A.K.; Hillman, L.S.; Peterson, C.A. Correcting vitamin D insufficiency improves insulin sensitivity in obese adolescents: A randomized controlled trial. Am. J. Clin. Nutr. 2013, 97, 774–781.
  41. Davidson, M.B.; Duran, P.; Lee, M.L.; Friedman, T.C. High-dose vitamin D supplementation in people with prediabetes and hypovitaminosis D. Diabetes Care 2013, 36, 260–266.
  42. Mitri, J.; Dawson-Hughes, B.; Hu, F.B.; Pittas, A.G. Effects of vitamin D and calcium supplementation on pancreatic beta cell function, insulin sensitivity, and glycemia in adults at high risk of diabetes: The Calcium and Vitamin D for Diabetes Mellitus (CaDDM) randomized controlled trial. Am. J. Clin. Nutr. 2011, 94, 486–494.
  43. Von Hurst, P.R.; Stonehouse, W.; Coad, J. Vitamin D supplementation reduces insulin resistance in South Asian women living in New Zealand who are insulin resistant and vitamin D deficient—A randomised, placebo-controlled trial. Br. J. Nutr. 2010, 103, 549.
  44. Jorde, R.; Sneve, M.; Torjesen, P.; Figenschau, Y. No improvement in cardiovascular risk factors in overweight and obese subjects after supplementation with vitamin D3 for 1 year. J. Intern. Med. 2010, 267, 462–472.
  45. Griffin, S.J.; Little, P.S.; Hales, C.N.; Kinmonth, A.L.; Wareham, N.J. Diabetes risk score: Towards earlier detection of type 2 diabetes in general practice. Diabetes. Metab. Res. Rev. 2000, 16, 164–171.
  46. Manson, J.E.; Cook, N.R.; Lee, I.-M.; Christen, W.; Bassuk, S.S.; Mora, S.; Gibson, H.; Gordon, D.; Copeland, T.; D’Agostino, D.; et al. Vitamin D Supplements and Prevention of Cancer and Cardiovascular Disease. N. Engl. J. Med. 2018, 380, 33–44.
  47. Rafiq, S.; Jeppesen, P.B. Body mass index, vitamin d, and type 2 diabetes: A systematic review and meta-analysis. Nutrients 2018, 10, 1182.
  48. Tang, H.; Li, D.; Li, Y.; Zhang, X.; Song, Y.; Li, X. Effects of Vitamin D Supplementation on Glucose and Insulin Homeostasis and Incident Diabetes among Nondiabetic Adults: A Meta-Analysis of Randomized Controlled Trials. Int. J. Endocrinol. 2018, 2018, 7908764.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 183
Revisions: 2 times (View History)
Update Date: 08 Aug 2023
1000/1000