Hostname: page-component-8448b6f56d-sxzjt Total loading time: 0 Render date: 2024-04-20T05:32:32.377Z Has data issue: false hasContentIssue false

Vitamin D and cardiometabolic health: a review of the evidence

Published online by Cambridge University Press:  01 December 2010

Siobhan Muldowney
Affiliation:
Vitamin D Research Group, School of Food and Nutritional Sciences, University College Cork, Republic of Ireland
Mairead Kiely*
Affiliation:
Vitamin D Research Group, School of Food and Nutritional Sciences, University College Cork, Republic of Ireland
*
*Corresponding author: Dr Mairead Kiely, fax +353 21 4270244, email m.kiely@ucc.ie
Rights & Permissions [Opens in a new window]

Abstract

The cardiometabolic syndrome (MetS) is a clustering of related metabolic abnormalities including abdominal adiposity, insulin resistance, hypertension, dyslipidaemia and increased inflammatory and thrombotic markers, which is linked to increased risk of type 2 diabetes, CVD and overall mortality. Several cross-sectional and prospective studies have shown an association between low vitamin D status, as indicated by concentrations of serum 25-hydroxyvitamin D (s25(OH)D), and increased prevalence of the MetS and individual CVD risk factors. These epidemiological observations are supported by mechanistic studies but experimental data are limited. The available data from intervention studies are largely confounded as most vitamin D supplementation trials were mainly carried out to explore the role of Ca in CVD and include Ca in the treatment arms. Inadequate consideration of seasonal effects on s25(OH)D concentrations is also a common design flaw in most studies. Further complications arise from shared risk factors such as adiposity and ageing, which predispose individuals to exhibit both a more pronounced risk profile and relatively lower s25(OH)D concentrations. In conclusion, while epidemiological associations are promising and a rationale for low vitamin D status as a potentially modifiable risk factor for CVD is supported by mechanistic data, suitable experimental data from appropriately designed trials are just beginning to emerge. As yet, this body of literature is too immature to draw firm conclusions on the role of vitamin D in CVD prevention. Carefully controlled vitamin D trials in well-described population groups using intervention doses that are titrated against target s25(OH)D concentrations could yield potentially valuable outcomes that may have a positive impact on CVD risk modification.

Type
Review Article
Copyright
Copyright © The Authors 2011

Introduction

CVD is a major cause of mortality and morbidity. In 2005, CVD accounted for 35·3 % of all deaths in the USA(Reference Lloyd-Jones, Adams and Carnethon1). Similarly, in Ireland, CVD resulted in 35 % of all deaths in 2006(2). Non-modifiable risk factors of CVD include increasing age, sex, family history and ethnicity(Reference Thom, Haase and Rosamond3), whereas modifiable risk factors include abnormal blood lipids (low HDL-cholesterol, apoB/apoA1), smoking, diabetes, high blood pressure, abdominal obesity, lack of physical exercise, diet, psychosocial stress and over-consumption of alcohol(Reference Yusuf, Hawken and Ounpuu4).

The cardiometabolic syndrome (MetS), first described by Kylin(Reference Kylin5), is defined as a clustering of metabolic abnormalities including obesity, high fasting blood glucose, insulin resistance, hypertension and dyslipidaemia, which are linked to increased risks of serious health complications such as type 2 diabetes, CVD, stroke and kidney failure. An estimated 47 million US residents have the MetS; the prevalence ranges from 6·7 % among young adults aged 20–29 years to 43·5 % among older adults aged 60–69 years and 42 % among those aged ≥ 70 years(Reference Ford, Giles and Dietz6). The prevalence of the MetS is 15 % in non-diabetic European adults over 30 years(Reference Hu, Qiao and Tuomilehto7) and this is expected to rise with increasing prevalence of obesity. In a subsample of middle-aged Irish men and women (n 1018; aged 50–69 years), Villegas et al. (Reference Villegas, Perry and Creagh8) estimated the prevalence of the MetS to be 21 % using the definition proposed by the WHO(9) or 20·7 % according to Third National Cholesterol Education Program Adult Treatment Panel (NCEP/ATP III) criteria(10).

Identification of the multiple genetic and environmental factors that predispose some overweight individuals to developing the various metabolic disorders that constitute the MetS would be a valuable step in helping to address the condition. In this context, nutrition, including vitamin D status, is potentially a modifiable risk factor. Numerous reports have linked low vitamin D status with an increased risk of the MetS and CVD risk(Reference Ford, Ajani and McGuire11, Reference Dobnig, Pilz and Scharnagl12). Therefore, given both the increasing prevalence of the MetS among adults and evidence of widespread low vitamin D status, exploration of these relationships could be a valuable step towards addressing one (or both) problems.

Scope of review and approach

The present review will describe epidemiological associations between vitamin D and cardiometabolic health. It will outline the mechanisms proposed for the association and will evaluate the recent experimental evidence. Searches were made on PubMed and ISI Web of Knowledgesm up to May 2010 using search terms to capture generic and specific words relevant to the topic, including ‘vitamin D’, ‘cholecalciferol’, ‘cardiovascular disease’, ‘metabolic syndrome’, ‘cardio-metabolic syndrome’, ‘blood pressure’, ‘hypertension’, ‘glucose’, ‘diabetes’, ‘insulin’, ‘insulin resistance’, ‘glucose metabolism’, ‘lipids’, ‘dyslipidemia’, ‘cholesterol’, ‘high-density lipoprotein’, ‘HDL’, ‘low-density lipoprotein’, ‘LDL’, ‘inflammation’, ‘C-reactive protein’, ‘CRP’, ‘interleukin-6’, ‘IL-6’, ‘tumour necrosis factor alpha’, ‘TNF-α’, ‘plasminogen activator inhibitor type 1’ (PAI-1), ‘matrix metalloproteinases’, ‘adhesion molecules’(ICAM-1 and VCAM-1), and combinations of these. We also manually searched bibliography lists of some primary articles for additional studies. The search on randomised controlled trials (RCT) was carried out from January 1990 to May 2010, and excluded RCT that used calcitriol (1,25-dihydroxyvitamin D (1,25(OH)2D)) or vitamin D analogues (for example, 1α-calcidiol). RCT using cholecalciferol or ergocalciferol, alone or in combination with Ca, were included; however, no RCT using ergocalciferol were identified in the time-frame specified, except for one which looked primarily at risk of falls. We restricted the search to articles in English.

Vitamin D endocrine system

The two most common forms of vitamin D are ergocalciferol (vitamin D2) and cholecalciferol (vitamin D3). Cholecalciferol is synthesised on skin exposure to UV blue (UVB) radiation. In the skin, 7-dehydrocholesterol is converted to precholecalciferol (pre-vitamin D), which is unstable. Through thermal isomerisation, pre-vitamin D is converted to cholecalciferol. Extra vitamin D3 can be obtained from the diet, although sources are limited and include oily fish, egg yolks and fortified foods. Ergocalciferol is produced by UV irradiation of the fungal steroid ergosterol and is used in some vitamin D supplements. Although the relative ability of vitamin D2 to increase vitamin D status (as indicated by serum concentrations of 25-hydroxyvitamin D (s25(OH)D)) compared with vitamin D3 was previously disputed, recent data from Holick et al. (Reference Holick, Biancuzzo and Chen13) showed that vitamin D2 was as effective as vitamin D3.

Both forms of vitamin D eventually enter the circulation and, bound to the vitamin D binding protein, are transported to the liver where the first of two hydroxylation steps occurs, producing the predominant circulating vitamin D metabolite, 25-hydroxyvitamin D (25(OH)D). Still bound to the vitamin D binding protein, 25(OH)D is transported around the body. The enzyme, 25-hydroxyvitamin D3-1-α-hydroxylase (CYP27B1), which is a member of the P450 family, is responsible for the second hydroxylation step, which produces 1,25(OH)2D, the active form of vitamin D. While CYP27B1 is located in many cells throughout the body, including smooth muscle cells, immune cells and pancreatic β-cells, the primary site of this hydroxylation step is in the mitochondria of the renal cortex, which appears to be tightly controlled by parathyroid hormone (PTH) for the maintenance of Ca homeostasis. However, non-renal CYP27B1 appears to operate outside this control(Reference Abdelkefi, Achour and Ben Othman14). The most accurate method of determining an individual's vitamin D status is to measure 25(OH)D concentrations in serum. Serum 25(OH)D circulates at about 1000 times the concentration of 1,25(OH)2D and has a longer half-life of approximately 2 weeks(Reference Holick15).

The action of vitamin D is achieved through a vitamin D receptor (VDR)-mediated mechanism in which 1,25(OH)2D regulates transcription of a wide variety of genes in vitamin D target cells(Reference Haussler, Whitfield and Haussler16). This mechanism involves forming a heterodimeric complex with a retinoic acid X receptor (1,25(OH)2D/VDR/RXR); once this complex binds with a vitamin D-responsive element it can exert various actions. In addition, 1,25(OH)2D can act through intra-cellular signalling pathways activated via plasma membrane receptors. The VDR has been identified in most body cells, including the small intestine, colon, brain, skin, prostate, gonads, breast, lymphocytes, macrophages, smooth muscle and β-cells(Reference Holick15), which has led to the hypothesis that vitamin D is implicated in the pathogenesis of many disorders, including certain types of cancer, diabetes (type 1 and type 2), CVD and several immune and inflammatory-related diseases(Reference Holick15, Reference Zittermann17).

Links between vitamin D and CVD

The story linking vitamin D and CVD has evolved substantially since the first studies, which date back to the 1960s. Data from animal studies suggested that hypervitaminosis D caused calcification and was involved in the pathogenesis of atherosclerosis(Reference Hass, Landerholm and Hemmens18Reference Morrison, Bajwa and Alfin-Slater20). In human subjects, Linden(Reference Linden21) showed evidence indicating that supplementary vitamin D was hypercholesterolaemic and increased the risk of myocardial infarction, and few studies found contradictory results(Reference Schmidt-Gayk, Goossen and Lendle22, Reference Vik, Try and Thelle23). Recent research in the field has proceeded along conventional lines, where mechanistic and experimental data seek to explore, explain, prove or dismiss epidemiological associations between low vitamin D status and indicators of metabolic dysfunction, the MetS and CVD.

With the increasing prevalence of obesity and a greater emphasis on the continuum which represents metabolic dysfunction and insulin resistance, with downstream progression to type 2 diabetes and eventually CVD, the research focus has shifted from specific studies on dyslipidaemia or hypertension to investigations of the interlinked metabolic irregularities which constitute the MetS. Several mechanisms are implicated in a putative role for vitamin D in CVD risk modification. Vascular smooth muscle contains VDR and CYP27B1, the enzyme necessary for the hydroxylation of 25(OH)D to 1,25(OH)2D, suggesting that vasculature is an important target tissue for vitamin D(Reference Zittermann and Koerfer24). Actions including suppression of vascular calcification, inhibition of vascular smooth muscle proliferation, modulation of inflammatory cytokines and regulation of the renin–angiotensin system (RAS) have all been described(Reference Zittermann, Schleithoff and Koerfer25).

To help the reader to weave through the large body of data available on this topic, we have taken a similar approach for each section of the present review: with respect to CVD, type 2 diabetes and the MetS as primary adverse health outcomes, as well as for contributing clinical risk factors, such as obesity, hypertension, dyslipidaemia, insulin resistance and inflammation, we have summarised epidemiological associations between the prevalence of each outcome or risk factor and vitamin D status, and followed this with an appraisal of the experimental and mechanistic data available.

CVD

It is interesting to note that the rate of CVD-related death is elevated at higher latitudes. However, data directly relating vitamin D status to the risk of CVD are meagre(Reference Grimes, Hindle and Dyer26). Using data from the Health Professionals Follow-up Study, Giovannucci et al. (Reference Giovannucci, Liu and Hollis27) showed that men (n 18 225) with a s25(OH)D < 37·5 nmol/l were at increased risk for myocardial infarction (RR 2·09; 95 % CI 1·24, 3·54) compared with those above 75 nmol/l after controlling for age, smoking status, BMI, family history, alcohol consumption, physical activity, history of diabetes mellitus and hypertension, ethnicity, marine n-3 intake, LDL- and HDL-cholesterol and TAG. Similarly, the Ludwigshafen Risk and Cardiovascular Health study reported an independent association of low s25(OH)D (median 19 nmol/l) with all-cause (hazard ratio (HR) 2·08; 95 % CI 1·60, 2·70) and cardiovascular mortality (HR 2·22; 95 % CI 1·57, 3·13) compared with high s25(OH)D (median 70·9 nmol/l)(Reference Dobnig, Pilz and Scharnagl12).

With respect to intervention studies, two large RCT have examined the effect of cholecalciferol (alone(Reference Trivedi, Doll and Khaw28) or in combination with Ca(Reference Hsia, Heiss and Ren29)) on CVD events. In the UK, Trivedi et al. (Reference Trivedi, Doll and Khaw28) showed no effect of 100 000 IU cholecalciferol every 4 months for 5 years (about 20 μg/d) on mortality from CVD in 2686 elderly adults (although it was suggestive of a reduced risk; RR 0·84; 95 % CI 0·65, 1·10; P = 0·2). It is important, however, to note that the primary outcome of this trial was fracture incidence, for which a protective effect of supplementation was noted. Hsia et al. (Reference Hsia, Heiss and Ren29) evaluated the risk of coronary and cerebrovascular events in the Women's Health Initiative trial of Ca plus vitamin D supplementation. This trial appears several times during the course of the present review. Briefly, 36 282 postmenopausal women aged between 50 and 79 years were randomised to receive either 1000 mg elemental Ca plus 10 μg cholecalciferol per d, or a matching placebo. After 7 years of follow-up, no effect on coronary or cerebrovascular risk was observed(Reference Hsia, Heiss and Ren29). Similarly, a more recent analysis of the Women's Health Initiative reported no difference in total and cause-specific mortality (including CVD and CHD deaths) in those who received the Ca and vitamin D supplementation compared with placebo(Reference LaCroix, Kotchen and Anderson30). There are a number of points to note with respect to the conduct of this trial and implications of the study outcomes; vitamin D and Ca supplementation was permitted among participants in addition to the randomisation protocol. Moreover, compliance was low, with approximately 60 % of participants adhering to an 80 % compliance rate. Lastly, habitual intakes of vitamin D and Ca were quite high (about 9·2 μg/d and about 1150 mg/d, respectively). A third RCT, albeit with falls prevention as the primary outcome, looked at the effect of intervening with ergocalciferol (25 μg/d) plus Ca (1000 mg/d) for 1 year in elderly women (aged 70–90 years) with a baseline s25(OH)D < 60 nmol/l(Reference Prince, Austin and Devine31). While the intervention reduced the risk of falls there was no effect on the risk of IHD or stroke. A recent meta-analysis(Reference Bolland, Avenell and Baron32) reviewing whether Ca supplementation increased the risk of myocardial infarction and other cardiovascular events in adults (aged>40 years) with, or at risk of osteoporosis, showed that Ca supplementation (>500 mg/d) increased the risk of myocardial infarction (HR 1·31; 95 % CI 1·02, 1·67). These data imply that giving Ca with vitamin D in RCT may obscure any benefits of vitamin D on reducing cardiovascular events and strengthens the need for vitamin D-only RCT to examine CVD incidence and mortality as a primary outcome.

Cardiometabolic syndrome

Several sets of definitive criteria are available to describe the MetS(9, 10, Reference Alberti, Zimmet and Shaw33Reference Grundy, Cleeman and Daniels35), which are used to guide therapeutic and clinical interventions as well as public health policy. The two most commonly used definitions are provided by the US National Cholesterol Education Program report(10) and the International Diabetes Federation(Reference Alberti, Zimmet and Shaw33). The two definitions are similar and will identify many of the same individuals as having the MetS. However, the International Diabetes Federation regards an increased waist circumference as an essential criterion for diagnosis, while the NCEP/ATP III requires the presence of any three criteria as sufficient, plus, while the International Diabetes Federation uses ethnic-specific cut-offs for waist circumference, the NCEP/ATP III use only one set of cut-offs, regardless of ethnicity.

The importance of identifying subjects with the MetS is because they are at increased risk of developing CVD, type 2 diabetes, stroke and kidney failure and in addition, each component can be treated on an individual basis. Hu et al. (Reference Hu, Qiao and Tuomilehto7), using data from eleven prospective European cohort studies, reported HR for cardiovascular mortality in adults with the MetS (defined by WHO criteria) of 2·26 (95 % CI 1·61, 3·17) and 2·78 (95 % CI 1·57, 4·94) in men and women, respectively, after controlling for age, blood cholesterol concentration and smoking. Clustering these risk factors has advantages over the analysis of the single entities by themselves. A prospective study, with a 6·9-year follow-up of first-degree relatives of patients with type 2 diabetes (n 4483), showed the relative risk (RR) for having CHD was greater in patients with the MetS (RR 2·96) compared with obesity (RR 1·44), dyslipidaemia (RR 1·73), hypertension (RR 1·57), insulin resistance (RR 1·53) and microalbuminuria (RR 0·94)(Reference Isomaa, Almgren and Tuomi36).

Low vitamin D status has been associated with an increased risk of the MetS (Ford et al. (Reference Ford, Ajani and McGuire11); Forouhi et al. (Reference Forouhi, Luan and Cooper37); Hypponen et al. (Reference Hypponen, Boucher and Berry38); Reis et al. (Reference Reis, von Muhlen and Miller39); S Muldowney, A Lucey, G Paschos, et al., unpublished results; M Kiely, S Muldowney, TR Hill, et al., unpublished results). Using data from National Health and Nutrition Examination Survey (NHANES) III in almost 8500 adults (aged ≥ 20 years), Ford et al. (Reference Ford, Ajani and McGuire11) observed subjects in the lowest quintile of s25(OH)D ( ≤ 48·4 nmol/l) were twice as likely to have the MetS compared with those in the highest quintile ( ≥ 96·4 nmol/l). Forouhi et al. (Reference Forouhi, Luan and Cooper37), using data from the Medical Research Council Ely Prospective Study, showed that in non-diabetic adults aged ≥ 40 years (n 524), baseline s25(OH)D was inversely associated with 10-year increased risk of the MetS. In a cross-sectional analysis of the 1958 British Birth Cohort, Hypponen et al. (Reference Hypponen, Boucher and Berry38) also showed in 6810 adults aged 44–46 years that s25(OH)D concentration was inversely associated with the prevalence of the MetS. Both of these studies showed interactions between s25(OH)D and insulin-like growth factor (IGF)-1; in the 1958 birth cohort, risk of the MetS was lowest among those with highest levels of both s25(OH)D and IGF-1(Reference Hypponen, Boucher and Berry38) whereas in the Ely cohort(Reference Forouhi, Luan and Cooper37), 25(OH)D increased with increasing IGF-1, which, in those with high IGF-1, was inversely related to IGF binding protein-1 and 2 h glucose concentration. Further analysis of the mechanisms by which 25(OH)D and the IGF-1 and IGF binding proteins interact is warranted to discover their influence on insulin and glucose secretion and/or sensitivity. Another recent cross-sectional analysis of 1654 men and women reported a substantially lower OR for the MetS in the highest quintile of s25(OH)D (median 88·0 nmol/l; OR 0·15) compared with the lowest quintile (median 26·8 nmol/l; OR 0·46)(Reference Reis, von Muhlen and Miller39). Recently, we have shown in young overweight adults that those with a s25(OH)D < 42·5 nmol/l were more likely to have the MetS (OR 3·46; 95 % CI 1·07, 11·18) compared with subjects with a s25(OH)D>63 nmol/l (S Muldowney, A Lucey, G Paschos, et al., unpublished results). Similarly, we showed in a representative sample of 12- and 15 year-old adolescents that the risk of having the MetS was increased (OR 3·47; 95 % CI 0·93, 13·01) in adolescents with a s25(OH)D < 48·2 nmol/l compared with those who had a s25(OH)D>74·7 nmol/l (M Kiely, S Muldowney, TR Hill, et al., unpublished results). However, not all studies have observed this association(Reference Hjelmesaeth, Hofso and Aasheim40Reference Reis, von Muhlen and Kritz-Silverstein42). McGill et al. (Reference McGill, Stewart and Lithander41) showed in 243 obese adults, 80 % of whom were women (age 47·6 (sd 12) years), that while s25(OH)D concentration was inversely associated with waist circumference, BMI and HbA1c, there was no association with the MetS. Using data from the Rancho Bernardo Study (n 1070; aged 44–96 years), Reis et al. (Reference Reis, von Muhlen and Kritz-Silverstein42) showed no association between s25(OH)D and MetS risk despite the strong positive association between PTH and MetS risk (OR 2·02; 95 % CI 0·96, 4·24). In this latter study mean s25(OH)D concentration was high (men, 108·9 nmol/l; women, 101·6 nmol/l) which may hide an association, if apparent. A recent systematic review and meta-analysis, using data from eight cross-sectional studies, showed that the prevalence of the MetS was reduced by approximately 50 % (OR 0·49; 95 % CI 0·38, 0·64) by having high s25(OH)D concentration(Reference Parker, Hashmi and Dutton43).

Glucose and insulin metabolism and type 2 diabetes

Impaired glucose-mediated insulin production and/or insulin resistance are key components of the MetS. Defects in pancreatic β-cell function, insulin sensitivity, and systemic inflammation are often present before glucose intolerance and/or type 2 diabetes develop(Reference Hu, Qiao and Tuomilehto7, Reference Weyer, Bogardus and Pratley44). There are several potential mechanisms by which vitamin D may influence glucose tolerance, and insulin release or sensitivity(Reference Pittas, Lau and Hu45, Reference Teegarden and Donkin46).

Firstly, pancreatic β-cells contain VDR and express 1α-hydroxylase(Reference Bland, Markovic and Hills47). Zeitz et al. (Reference Zeitz, Weber and Soegiarto48) reported that circulating glucose concentration was higher and insulin concentration was lower in mice with non-functioning VDR compared with wild-type mice. Local production of 1,25(OH)2D enhances the synthesis and release of insulin by β-cells in response to glucose stimulation(Reference Bourlon, Billaudel and Faure-Dussert49). Secondly, calbindin, a vitamin D-dependent Ca-binding protein, has long been known to be present in the pancreas of various species, including humans(Reference Arnold, Kuttner and Swaminathan50, Reference Morrissey, Bucci and Richard51). Sooy et al. (Reference Sooy, Schermerhorn and Noda52) showed that calbindin is an important regulator of insulin production, by regulating Ca concentration in the β-cell. Thirdly, s25(OH)D concentration was related to insulin sensitivity in a group of healthy non-diabetic adults(Reference Chiu, Chu and Go53), and a recent animal study demonstrated that while 1,25(OH)2D treatment had no effect on insulin receptor expression in non-diabetic rats, it normalised the number of insulin receptors and improved insulin response to glucose transport in adipocytes in streptozotocin-induced diabetic rats, most probably by direct transcriptional activation of the rat insulin receptor gene(Reference Calle, Maestro and Garcia-Arencibia54).

Elevated PTH concentrations have been shown to be inversely associated with insulin sensitivity in healthy adults(Reference Chiu, Chuang and Lee55), possibly through its effects on intracellular free Ca concentration of the pancreatic β-cell, adipocytes or muscle cells(Reference McCarty and Thomas56). In some instances, PTH and not vitamin D has been associated with glucose tolerance(Reference Wareham, Byrne and Carr57); however, other studies have found no association(Reference Hjelmesaeth, Hofso and Aasheim40, Reference Snijder, van Dam and Visser58).

VDR gene polymorphisms may also have a role to play. Several polymorphisms in the VDR gene have been described, with Apa I, Bsm I, Taq I and Fok I being the most commonly investigated in relation to type 2 diabetes, glucose homeostasis and insulin release(Reference Reis, Hauache and Velho59, Reference Palomer, Gonzalez-Clemente and Blanco-Vaca60). Numerous case–control studies have found no difference in the distribution of VDR polymorphisms between diabetic patients and control subjects(Reference Dilmec, Uzer and Akkafa61Reference Malecki, Frey and Moczulski63); however, an association with a specific aspect, such as glucose intolerance or insulin secretion has been observed frequently(Reference Hitman, Mannan and McDermott64Reference Filus, Trzmiel and Kuliczkowska-Plaksej69). Hitman et al. (Reference Hitman, Mannan and McDermott64) showed a strong association between higher insulin secretion and Apa I VDR polymorphism (homozygous for the A allele) in healthy Bangladeshi adults living in London and at risk of type 2 diabetes, independent of vitamin D status. An extension to this study showed that VDR polymorphisms of Taq I in all subjects and Taq I and Fok I in subjects with s25(OH)D < 50 nmol/l were independent determinants of insulin secretion(Reference Ogunkolade, Boucher and Prahl65). Using data from The Rancho Bernardo Study, Oh & Barrett-Connor(Reference Oh and Barrett-Connor66) reported that fasting plasma glucose and prevalence of glucose intolerance were lower in non-diabetic individuals with the AA genotype compared with those with the aa genotype. This study also observed that subjects with the bb genotype of the Bsm I VDR polymorphisms were more likely to be insulin resistant compared with BB carriers, which contrasts with a more recent study where no association was observed between the Bsm I VDR polymorphism and insulin resistance(Reference Tworowska-Bardzinska, Lwow and Kubicka67). These contrasting results should be further explored, including possible interactions with potential confounders such as IGF-1 and its binding proteins(Reference Forouhi, Luan and Cooper37, Reference Hypponen, Boucher and Berry38), as well as PTH(Reference Chiu, Chuang and Lee55, Reference Wareham, Byrne and Carr57).

Other potential mechanisms may include effects through inflammatory processes(Reference Tai, Need and Horowitz70, Reference Timms, Mannan and Hitman71). Systemic inflammation is up-regulated in type 2 diabetic patients(Reference Hu, Qiao and Tuomilehto7, Reference Duncan, Schmidt and Pankow72, Reference Pradhan, Manson and Rifai73). While systemic inflammation has mainly been implicated in insulin resistance, elevated cytokines may initiate β-cell apoptosis. Vitamin D, through its inhibitory effect on pro-inflammatory cytokines(Reference Giulietti, van Etten and Overbergh74), may improve insulin sensitivity and encourage β-cell survival(Reference Pittas, Lau and Hu45). Interactions between vitamin D and inflammatory processes are described in more detail below.

Clinical evidence – observational studies

A high dietary intake of vitamin D and/or Ca, and dairy foods has been inversely associated with glucose and insulin concentration and incidence of type 2 diabetes(Reference Choi, Willett and Stampfer75Reference Pittas, Dawson-Hughes and Li78). Using data from the Women's Health Study, Liu et al. (Reference Liu, Song and Ford76) reported that a total vitamin D intake above about 13 μg/d was associated with a lower risk of incident type 2 diabetes compared with intakes below 4 μg/d (P = 0·02). Another prospective study, using data from the first Nurses’ Health Study (n 83 779), reported that women who consumed ≥ 20 μg/d of vitamin D had a 23 % lower risk of developing type 2 diabetes compared with women who consumed ≤ 5 μg vitamin D/d after controlling for BMI, age, physical activity, alcohol and caffeine use, family history of diabetes, smoking, hypertension and state of residence(Reference Pittas, Dawson-Hughes and Li78) (additional adjustment for other dietary factors removed significance). These effects reflected intakes from supplements; women who obtained ≥ 10 μg/d vitamin D from supplements had a 13 % lower risk of type 2 diabetes than women who took ≤ 2·5 μg/d, after adjusting for the same covariates plus dietary factors including Ca. This prospective study showed the effects of Ca and vitamin D in combination, in that women with the highest vitamin D ( ≥ 20 μg/d) and highest Ca intakes (1200 mg/d) had the lowest risk of diabetes (RR 0·67; 95 % CI 0·49, 0·90).

Using data from NHANES III, Scragg et al. (Reference Scragg, Sowers and Bell79) reported that s25(OH)D concentration was inversely related to the presence of type 2 diabetes and increased insulin resistance in US adults (aged 40–74 years); the OR for diabetes was 0·25 (95 % CI 0·11, 0·60) in non-Hispanic whites with s25(OH)D concentration ≥ 81 nmol/l compared with those ≤ 43·9 nmol/l. A cross-sectional analysis of the Framingham Offspring Study in 808 non-diabetic subjects (mean age 59·6 (sd 0·3) years) found that plasma 25(OH)D concentration (mean 47·4 (sd 0·6) nmol/l) was inversely associated with fasting plasma glucose, insulin and the homeostasis model assessment of insulin resistance (HOMA-IR, an indicator of insulin resistance), but not 2 h post-oral glucose tolerance test glucose, after adjusting for age, sex, BMI, waist circumference and current smoking habits(Reference Liu, Meigs and Pittas80). Subjects in the top third of vitamin D status (s25(OH)D ≥ 53·4 nmol/l) had a 1·6 % lower concentration of fasting plasma glucose (P for trend = 0·007), a 9·8 % lower concentration of fasting plasma insulin (P for trend = 0·001) and a 12·7 % lower HOMA-IR score (P for trend < 0·001) compared with subjects in the bottom third of vitamin D status (s25(OH)D ≤ 38·6 nmol/l). The HOMA-IR was only decreased in subjects with a plasma 25(OH)D concentration>81 nmol/l. In the Medical Research Council Ely Prospective Study, Forouhi et al. (Reference Forouhi, Luan and Cooper37) reported that s25(OH)D concentrations at baseline were inversely related with 10-year risk of hyperglycaemia, insulin resistance and the MetS. In a systematic review by Pittas et al. (Reference Pittas, Lau and Hu45) using data from four studies(Reference Wareham, Byrne and Carr57, Reference Snijder, van Dam and Visser58, Reference Scragg, Sowers and Bell79, Reference Scragg, Holdaway and Singh81), a summary OR for the association between s25(OH)D and type 2 diabetes of 0·54 (95 % CI 0·23, 1·27) was reported for the highest s25(OH)D concentration (62·5–95 nmol/l) v. the lowest (25–57·5 nmol/l).

There are three possible mechanisms for the development of the chronic complications of diabetes, glycation, polyol pathway and oxidative stress. Naturally, Hb is glycated to some extent (5–10 %), but this is increased (2- to 3-fold) in diabetics. Glycated Hb (most commonly HbA1c) levels are used as an index of recent blood glucose control. Inverse associations have been observed between s25(OH)D concentration and HbA1c(Reference Hypponen, Boucher and Berry38, Reference Scragg, Holdaway and Singh81), which may be more marked in obese subjects(Reference Hypponen and Power82).

Intervention studies

Data from RCT are relatively rare and to the best of our knowledge have been limited to patients with type 2 diabetes or other underlying diseases (for example, uraemia), patients who are overweight, insulin resistant or vitamin D deficient. RCT with cholecalciferol (as a single treatment; see Table 1) improved insulin response to glucose in some(Reference Nagpal, Pande and Bhartia83Reference von Hurst, Stonehouse and Coad85), but not all studies(Reference Jorde and Figenschau86). In India, Nagpal et al. (Reference Nagpal, Pande and Bhartia83) showed that large doses of vitamin D3 (three × 3000 μg, fortnightly) improved postprandial insulin sensitivity after 6 weeks in centrally obese healthy men (aged ≥ 35 years). Female type 2 diabetics, the vast majority of whom were vitamin D deficient, had improved first-phase insulin secretion and insulin resistance after supplementation with 33 μg vitamin D3 per d for 1 month(Reference Borissova, Tankova and Kirilov84). In a more long-term RCT, insulin sensitivity, insulin resistance and fasting insulin concentration were improved after 6 months of supplementation with 100 μg vitamin D3/d in South Asian women (n 81) who were insulin resistant (HOMA-IR ≥ 1·93) and vitamin D deficient (s25(OH)D < 50 nmol/l) at baseline(Reference von Hurst, Stonehouse and Coad85). In contrast, thirty-six type 2 diabetic patients, without vitamin D deficiency, who were randomised to receive 142·9 μg vitamin D3/d or placebo for 6 months, showed no improvement in glycaemic control(Reference Jorde and Figenschau86). Recently we completed an RCT in healthy adults, aged 20–40 years and ≥ 64 years, and showed that 15 μg vitamin D3/d offset the seasonal increase in fasting glucose concentration in the younger adults (S Muldowney, TR Hill, AJ Lucey, et al., unpublished results).

Table 1 The effect of cholecalciferol supplementation (as a single treatment) on cardiometabolic biomarkers

M, male; F, female; YA, young adults; OA, older adults; RDBPCT, randomised, double-blind, placebo-controlled trial; FG, fasting glucose; HOMA, homeostasis model assessment; IR, insulin resistance; SBP, systolic blood pressure; DBP, diastolic blood pressure; CHOL, cholesterol; CRP, C-reactive protein; s25(OH)D, serum 25-hydroxyvitamin D; T2DM, type 2 diabetes mellitus; IS, insulin sensitivity; OGIS, oral glucose insulin sensitivity; OGTT, oral glucose tolerance test; ICU, intensive care unit; RCT, randomised controlled trial; i.v., intravenously; i.m., intramuscularly; MMP, matrix metalloproteinase; TIMP, tissue inhibitor of metalloproteinases.

* Unpublished study by Muldowney S, Hill TR, Lucey AJ, et al. titled ‘Effect of vitamin D supplementation on biomarkers of CVD risk in 20–40 and 64+year old men and women’.

Another reason why RCT are difficult to interpret is that they have been confounded by the combination of Ca with vitamin D; therefore, in these studies it is not possible to be confident that the response observed is due to the vitamin D and/or Ca. Additionally, if the findings of the recent meta-analysis(Reference Bolland, Avenell and Baron32) with regard to cardiovascular events are correct, giving Ca with vitamin D may occlude the possible beneficial effects of vitamin D. Pittas et al. (Reference Pittas, Harris and Stark87) completed a post hoc analysis of 314 healthy adults, aged ≥ 65 years, who had originally completed a double-blind, placebo-controlled trial designed to assess skeletal outcomes, where subjects received either 500 mg calcium carbonate and 17·5 μg vitamin D3 or a placebo daily for 3 years(Reference Dawson-Hughes, Harris and Krall88). Positive effects of the supplementation compared with placebo on fasting plasma glucose and HOMA-IR were only apparent in subjects who had impaired fasting glucose at baseline (fasting plasma glucose ≥ 5·6 mmol/l). In a large double-blind RCT (n 330) in Norway, no effect of 500 mg Ca daily plus vitamin D3 (500 or 1000 μg/week) was shown in overweight and obese (>28–47 kg/m2) subjects on glucose metabolism after 1 year(Reference Jorde, Sneve and Torjesen89). Similarly, using data from the Women's Health Initiative, de Boer et al. (Reference de Boer, Tinker and Connelly90) showed that the incidence of diabetes at age 7 years did not differ between those receiving Ca and vitamin D compared with those receiving the placebo and there was no difference in fasting plasma glucose, insulin or HOMA-IR. Thus, the association between s25(OH)D concentrations and altered glucose and insulin metabolism reported widely in cross-sectional and prospective studies requires confirmation by RCT using vitamin D only.

Obesity

The prevalence of obesity is increasing worldwide, most probably due to positive energy balance associated with increasing sedentary lifestyles. The WHO estimated that 1·6 billion adults were overweight and 400 million adults were obese worldwide in 2005(91). In the USA, age-adjusted prevalence of obesity increased from 22·9 % in NHANES III (1988–94) to 30·5 % in 1999–2000(Reference Flegal, Carroll and Ogden92). Using data from the North/South Ireland Food Consumption Survey, McCarthy et al. (Reference McCarthy, Harrington and Kiely93) reported that 39 % of Irish adults were overweight and 17·8 % were obese, which compared with data from the Irish Nutrition Survey, 1990(Reference Lee and Cunningham94), is nearly a two-fold increase in obesity in 10 years (1990–2000). In recent years, the evidence for excess body weight as a causative agent in the MetS has encouraged the use of an adiposity measure as an essential element of defining the MetS(Reference Alberti, Zimmet and Shaw33).

Numerous studies have observed that obesity is a negative predictor of vitamin D status (for a review, see Pérez-López(Reference Pérez-López95)). For example, data from the sixth Tromso study, which was a longitudinal analysis(Reference Jorde, Sneve and Emaus96), showed an inverse association between s25(OH)D concentration and BMI in over 10 000 adults. Hypponen & Power(Reference Hypponen and Power82), using data from the 1958 British birth cohort (n 7189), observed that 80 % of subjects (BMI>30 kg/m2) had a s25(OH)D < 75 nmol/l, compared with 68 % of the non-obese subjects. In another study on 243 ambulant adults (BMI 28–50 kg/m2) enrolled in a weight-loss study, s25(OH)D concentration was inversely associated with weight (P = 0·0009), BMI (P = 0·005) and waist circumference (P = 0·03), but not body fat percentage(Reference McGill, Stewart and Lithander41). This study also estimated a decrease of 0·74 nmol/l in s25(OH)D per 1 kg/m2 increase in BMI.

Compston et al. (Reference Compston, Vedi and Ledger97) suggested that the inverse association between s25(OH)D and adiposity could be explained by the observation that obese individuals spend less time outdoors and are therefore exposed to less UVB radiation. However, a recent cross-sectional analysis of 385 healthy adults, aged ≥ 65 years, showed that sun exposure did not explain the inverse association between s25(OH)D levels and adiposity(Reference Harris and Dawson-Hughes98). Bell et al. (Reference Bell, Epstein and Greene99) proposed that the vitamin D endocrine system is altered in obese subjects, with increased production of 1,25(OH)2D exerting negative feedback control on the hepatic synthesis of 25(OH)D. In addition, animal studies have shown that 1,25(OH)2D increases lipogenesis and decreases lipolysis(Reference Shi, Norman and Okamura100, Reference Zemel, Shi and Greer101), and, therefore, may contribute to obesity; however, 1,25(OH)2D concentration is not consistently higher in obese subjects(Reference Parikh, Edelman and Uwaifo102, Reference Konradsen, Ag and Lindberg103). Furthermore, 1,25(OH)2D is not consistently related to 25(OH)D status. The efficiency of epidermal vitamin D production does not appear to be impaired in obesity, but adipose tissue appears to sequester cholecalciferol and 25(OH)D, making it unavailable to the circulation; thus obese individuals frequently have low s25(OH)D concentrations(Reference Wortsman, Matsuoka and Chen104). Several years ago, Arunabh et al. (Reference Arunabh, Pollack and Yeh105) concluded that body fatness was inversely related to s25(OH)D levels in healthy women, but more recently, Freedman et al. (Reference Freedman, Wagenknecht and Hairston106) confirmed a strong inverse association between 25(OH)D and visceral adiposity in African-Americans with diabetes. This latest finding has important implications for study design; if visceral fat is a potentially more powerful determinant of s25(OH)D levels in overweight individuals as well as a prognostic indicator of metabolic dysfunction, it may be more appropriate to measure visceral fat rather than overall body fatness.

Intervention studies

There is a plausible explanation to why vitamin D may have a role in weight management; however, the evidence is limited, as most RCT use vitamin D in combination with Ca. In Norway, 334 overweight/obese subjects were randomised to receive 500 mg Ca daily plus vitamin D3 (500 or 1000 μg/week) or placebo for 1 year(Reference Sneve, Figenschau and Jorde107). However, there was no change in weight throughout the intervention. Using data from the Women's Health Initiative, Caan et al. (Reference Caan, Neuhouser and Aragaki108) observed that postmenopausal women randomised to receive Ca and vitamin D3 had smaller average annual weight gain compared with the women taking the placebo; moreover, the favourable effect was strongest among women who had, at baseline, a Ca intake < 1200 mg/d. Another RCT showed healthy, overweight and obese women (n 63), partaking in a 15-week energy restriction ( − 700 kcal/d; 2930 kJ/d) intervention, who were randomised to receive a daily supplement of 10 μg vitamin D3 and 1200 mg Ca, improved their blood lipid profile compared with women randomised to receive a placebo(Reference Major, Alarie and Dore109); however, no difference in weight loss was observed between treatment groups. A reanalysis of this study, limited to women with a very low Ca intake (baseline Ca < 600 mg/d), showed that women receiving the combined vitamin D and Ca supplement had greater weight loss compared with those receiving the placebo(Reference Major, Alarie and Dore110). A more recent study investigated the effects of vitamin D supplementation (83 μg/d) for 1 year on weight loss and cardiovascular risk factors in overweight subjects who were participating in a weight-loss programme(Reference Zittermann, Frisch and Berthold111). Weight loss was similar in both treatment groups; however, combined vitamin D supplementation with weight loss resulted in a more pronounced improvement in several CVD risk markers than did weight loss alone. This RCT will be described later in the review. On the basis of this evidence, it is impossible to say whether vitamin D has any direct effect on the maintenance of a healthy body weight.

Elevated blood pressure

Elevated blood pressure is a common and independent risk factor for CVD, including stroke, heart failure and CHD. The seventh report of the Joint National Committee on prevention, detection, evaluation, and treatment of high blood pressure provides the most recent US classification of blood pressure levels(Reference Chobanian, Bakris and Black112). Using data from NHANES (1999–2000), Wang & Wang(Reference Wang and Wang113) reported that 27 % of US adults (n 4805; aged ≥ 18 years) had hypertension. The 2007 Survey of Lifestyle, Attitudes and Nutrition (SLÁN) showed in 1207 Irish adults (aged ≥ 45 years) that the prevalence of hypertension was 60 %(Reference Morgan, McGee and Watson114). Hypertension can be treated with antihypertensive medication and improved by changing modifiable risk factors such as body weight, smoking, physical activity and diet. Dietary approaches to reduce blood pressure have focused on the role of Na, fruit and vegetables and Ca. Growing evidence indicates that vitamin D may have an association with blood pressure, although the data are not consistent(Reference Pilz, Tomaschitz and Ritz115).

The role of 1,25(OH)2D in the RAS and its association with PTH are potential mechanisms by which vitamin D and blood pressure are linked. The RAS, also known as the renin–angiotensin–aldosterone system, helps regulate blood pressure, electrolyte levels, and volume homeostasis, and excessive RAS stimulation is associated with hypertension. Renin, a protease, primarily synthesised in the juxtaglomerular cell of the kidney, catalyses the conversion of angiotensinogen to angiotensin I, which is further cleaved to angiotensin II by angiotensin-converting enzyme in the capillaries of the lungs. Elevated angiotensin II raises blood pressure in one of two ways: (1) angiotensin II is a potent vasoconstrictor(Reference Ballermann and Marsden116) and elevates blood pressure by increasing vascular resistance; (2) angiotensin II stimulates aldosterone secretion, which in turn, increases Na reabsorption in the kidney and therefore blood pressure is raised.

Circulating 1,25(OH)2D has been shown to play a role in the regulation of the RAS; for example, 1,25(OH)2D was negatively correlated (r − 0·65; P < 0·001) with plasma renin activity(Reference Resnick, Muller and Laragh117). In this study, 1,25(OH)2D was elevated in hypertensive subjects with low renin activity but not normotensive or hypertensive subjects with high renin activity. Renin expression and angiotensin II production are elevated in VDR knock-out mice(Reference Li, Kong and Wei118). However, the expression of angiotensinogen did not differ between VDR knock-out and wild-type mice and this indicates that the elevated production of angiotensin II is primarily due to the increase in renin activity. The observed hypertension, cardiac hypertrophy and increased water intake were corrected by treatment with an angiotensin-converting enzyme inhibitor (captopril) or an angiotensin II AT1 receptor antagonist (losartan), confirming that it was the RAS responsible for the observed abnormalities. In addition, that study showed that inhibition of 1,25(OH)2D led to up-regulation of renin and, furthermore, treatment with 1,25(OH)2D suppressed renin expression in wild-type mice. A continuation of this study, using As4·1 cells, which are the best available in vitro model for a juxtaglomerular cell, showed that 1,25(OH)2D directly suppresses renin gene transcription, through a VDR-mediated mechanism. These results were supported by a second animal study(Reference Xiang, Kong and Chen119), where VDR knock-out mice developed excessive concentrations of renin in their blood, which led to high blood pressure. The molecular effects are still unknown but appear to be independent of both Ca and PTH concentration(Reference Xiang, Kong and Chen119Reference Zhou, Lu and Cao121). Instead, cyclic AMP, which is a major intracellular signal that stimulates renin expression, has been implicated. Yuan et al. (Reference Yuan, Pan and Kong122) showed that liganded VDR suppresses renin expression by binding to the transcription factor cyclic AMP-response element-binding protein. This inhibits renin transcription, as cyclic AMP-response element-binding protein is no longer available for binding to the cyclic AMP response elements in the promoter region of the renin gene.

Besides its role in Ca homeostasis, PTH has a pro-sclerotic effect on vascular smooth muscle cells(Reference Zemel123) which may contribute to vessel-wall thickening and subsequent rises in blood pressure. Elevated circulating levels of 1,25(OH)2D, as a result of increased PTH, stimulates Ca influx in a variety of cells, including the vascular smooth muscle cells, which results in contraction and increased vascular resistance, i.e. elevated blood pressure(Reference Perkovic, Hewitson and Kelynack124).

Clinical evidence – observational studies

The first indication that vitamin D was related to blood pressure in humans was from observations that residents at higher latitudes, who experience decreased UVB exposure as a result, had higher systolic and diastolic blood pressure (SBP and DBP)(Reference Rostand125). In fact, UVB light was shown, in addition to increasing s25(OH)D concentration, to lower blood pressure in adults (aged 26–66 years) with essential hypertension(Reference Krause, Buhring and Hopfenmuller126).

Cross-sectional and prospective studies examining the relationship between vitamin D intake and blood pressure have reported conflicting results(Reference Sowers, Wallace and Lemke127, Reference Jorde and Bonaa128); however, the evidence appears to be strongest for SBP. Sowers et al. (Reference Sowers, Wallace and Lemke127) examined eighty-six normotensive young women (aged 20–35 years) and 222 older normotensive women (aged 55–80 years) to investigate if Ca and vitamin D intakes affected bone density or blood pressure. In the young women, vitamin D intake above 10 μg/d was predictive of a lower SBP ( − 6 mmHg) but no association was apparent with DBP. In the older women, having an intake of both vitamin D and Ca above 10 μg and 800mg/d, respectively, was predictive of lower SBP. In a larger, more recent study of over 15 000 generally healthy Norwegians (aged 25–69 years) who took part in the fourth Tromso study, no association between vitamin D intake and blood pressure was observed after multivariate adjustment(Reference Jorde and Bonaa128); however, very few subjects had a vitamin D intake>10 μg/d. Results from three large prospective studies, Nurses' Health Study I (n 77 436), Nurses' Health Study II (n 93, 803) and the Health Professionals Follow-up Study (n 38 074), found no evidence that a higher intake of vitamin D reduced (or increased) the risk of incident hypertension after multiple adjustments and approaches to the analysis(Reference Forman, Bischoff-Ferrari and Willett129). Diagnosis of hypertension in this study was self-reported, and although self-reported hypertension in a health professionals' study is reasonably reliable, it does cast doubt over the study outcomes. Data from the Women's Health Study found that intakes of low-fat dairy products, Ca, and vitamin D were each inversely associated with risk of hypertension in middle-aged and older women(Reference Wang, Manson and Buring130).

The association between s25(OH)D concentration and blood pressure is also inconsistent. Pasco et al. (Reference Pasco, Henry and Nicholson131) showed in a sample of Australian women (n 861; aged 20–92 years) that the odds for having a high blood pressure was reduced in those with a s25(OH)D in the top or middle third compared with those in the lowest third (median 40 nmol/l), after accounting for weight and other potential confounders. Additionally, we recently reported that adolescents with a s25(OH)D < 60·1 nmol/l were more likely (OR 1·94; 95 % CI 1·18, 3·20) to have an elevated SBP (>135 mmHg) compared with those who had a s25(OH)D>60·1 nmol/l (M Kiely, S Muldowney, TR Hill, et al., unpublished results). In contrast, Snijder et al. (Reference Snijder, Lips and Seidell132) using data from the Longitudinal Aging Study Amsterdam observed no association between 25(OH)D concentration and blood pressure in 1205 elderly subjects; however, 80 % of these subjects had hypertension. Similarly, using data from the fourth and sixth Tromso Study, Jorde et al. (Reference Jorde, Figenschau and Emaus133) showed that after adjustment for confounding, s25(OH)D did not predict future hypertension risk or increase in blood pressure. Three articles using data from NHANES III have reported on the association between vitamin D and blood pressure/hypertension(Reference Scragg, Sowers and Bell134Reference Martins, Wolf and Pan136). Scragg et al. (Reference Scragg, Sowers and Bell134) showed that after multivariate adjustment for sex, age, ethnicity and physical activity, mean SBP and DBP were lower in the highest quintile of vitamin D status (s25(OH)D>85·7 nmol/l) than in the lowest quintile (s25(OH)D < 40·4 nmol/l). Judd et al. (Reference Judd, Nanes and Ziegler135) observed an inverse association between 25(OH)D and SBP in white, but not black, men and women; however, adjustment for age removed the association. In addition, this study showed that in white, but not black, subjects a s25(OH)D>80 nmol/l reduced the age-related increase in SBP by 20 % compared with white subjects with s25(OH)D < 50 nmol/l, taking BMI, sex, physical activity and smoking status into consideration. Further analysis of NHANES III indicates that having a s25(OH)D concentration < 52·5 nmol/l is associated with an increased risk of hypertension (OR 1·30; 95 % CI 1·13, 1·49) compared with having a 25(OH)D ≥ 92·5 nmol/l(Reference Martins, Wolf and Pan136). The confounding effects of age and ethnicity in cross-sectional studies could overpower any putative associations between 25(OH)D levels and blood pressure.

The question of whether low vitamin D status is a risk factor for the development of hypertension has been addressed by prospective studies. Using data from the Health Professionals Follow-up Study (n 613) and the Nurses' Health Study I (n 1198), Forman et al. (Reference Forman, Giovannucci and Holmes137) showed that plasma 25(OH)D concentration was inversely associated with risk of incident hypertension in men and women, after 8 years of follow-up accounting for age, BMI, physical activity, race, menopausal status (for women) and other potential confounders. In analysis of the pooled data (Health Professionals Follow-up Study and Nurses' Health Study I), after 4 years of follow-up, individuals whose 25(OH)D levels were < 37·5 nmol/l compared with those whose levels were ≥ 75 nmol/l had an RR for incident hypertension of 3·18 (95 % CI 1·39, 7·29). Using data from the Nurses' Health Study II, Forman et al. (Reference Forman, Curhan and Taylor138) observed that women who were vitamin D deficient (25(OH)D < 75 nmol/l) had a 47 % increased risk (OR 1·47; 95 % CI 1·10, 1·97) of incident hypertension after adjustment compared with those who had 25(OH)D>75 nmol/l. This study estimated that vitamin D deficiency may account for 23·7 % of all new cases of hypertension developing among young women every year. Collectively, these data indicate that the apparent inverse association between vitamin D status and blood pressure and hypertension is of a sufficient magnitude to have a potentially important impact on cardiovascular mortality(Reference Scragg, Khaw and Murphy139).

Intervention trials

To date, only one RCT using cholecalciferol only has been completed with blood pressure as the primary outcome (see Table 1). Scragg et al. (Reference Scragg, Khaw and Murphy139) randomised elderly (n 189; aged 63–76 years) subjects to receive a single supra-physiological dose of vitamin D3 (2·5 mg) or a placebo during the winter; however, it had no effect on SBP or DBP. Unexpectedly, this study did report a decrease in radial pulse rate in subjects receiving the vitamin D, which is a crude measure of cardiac function. The lack of a treatment effect on SBP or DBP in this study may be due to the short follow-up time (5 weeks) but is more probably due to the effect of age itself on blood pressure in subjects who have had hypertension for some time. The vascular damage resulting from long-term hypertension makes older subjects with established hypertension unsuitable candidates for studies of vitamin D supplementation for blood pressure reduction. Some RCT looking at CVD risk factors, or trials more specific to insulin or glucose metabolism, have also reported blood pressure data (see Table 1). We recently used data from a RCT with cholecalciferol to explore its effects on biomarkers of CVD (S Muldowney, TR Hill, AJ Lucey, et al., unpublished results); however, no effect on blood pressure was apparent (in young or older healthy adults). All other vitamin D RCT that reported blood pressure data were in combination with Ca or used UVB therapy(Reference Jorde, Sneve and Torjesen89, Reference Krause, Buhring and Hopfenmuller126, Reference Orwoll and Oviatt140Reference Pfeifer, Begerow and Minne142).

In 1990, Orwoll & Oviatt(Reference Orwoll and Oviatt140) reported no effect of Ca (1000 mg/d) and cholecalciferol (25 μg/d) supplementation for 3 years on blood pressure in thirty-five men (aged 30–92 years) compared with thirty age-matched controls, but given the large age range, this study had insufficient power to detect a difference. Similarly, the Women's Health Initiative found no effect of Ca and cholecalciferol on blood pressure in postmenopausal women(Reference Margolis, Ray and Van Horn141). More recently, Jorde et al. (Reference Jorde, Sneve and Torjesen89) showed that overweight and obese subjects who were randomised to receive 500 mg Ca daily plus cholecalciferol (500 or 1000 μg/week) for 1 year had a slightly higher increase in SBP compared with those who received the Ca plus a placebo (2·3 v. − 0·1 mmHg); however, this study was powered to examine an effect on weight loss not blood pressure. On the other hand, Pfeifer et al. (Reference Pfeifer, Begerow and Minne142) randomised women, aged ≥ 70 years, who were vitamin D insufficient (s25(OH)D < 50 nmol/l) to receive daily either 1200 mg Ca or 1200 mg Ca plus 20 μg cholecalciferol for 8 weeks. SBP fell by 13·1 mmHg in women receiving the vitamin D plus Ca, whereas in women receiving Ca alone, SBP reduced by 5·1 mmHg. No difference in DBP was observed between the two treatment groups.

Mechanistic evidence is strong for an inverse association between vitamin D and blood pressure; however, the data available to date have added little support for a positive effect of vitamin D on blood pressure. A useful approach may be to select slightly younger adults without long-standing hypertension who have low vitamin D status for participation in RCT.

Dyslipidaemia

Cholesterol is an essential substance, synthesised in the body and obtained from the diet, and is necessary for maintaining cell membrane integrity and for the manufacture of hormones(Reference Cox, Garcia-Palmieri, Walker, Hall and Hurst143). Cholesterol and TAG are transported in the circulation on lipoproteins synthesised in the liver. LDL carries 60–80 % of the body's cholesterol, while HDL is involved in reverse cholesterol transport. Apoproteins facilitate cholesterol transport by providing recognition sites for cell-surface receptors and acting as cofactors for enzymes involved in the metabolism of lipoproteins. ApoA1 is the major apoprotein on HDL and is anti-atherogenic under most conditions. ApoB is associated with chylomicrons, VLDL (and their remnants) and LDL. All forms of apoB are potentially atherogenic if their metabolism is not regulated properly(Reference Cox, Garcia-Palmieri, Walker, Hall and Hurst143). Lipoprotein(a), a lipoprotein subclass, is found in variable concentrations and its levels are highly heritable. Lipoprotein(a) is a potential risk factor for CVD as it interferes with plasminogen and stimulates the production of PAI-1, thus reducing fibrinolysis and promoting clot formation(Reference Tziomalos, Athyros and Wierzbicki144).

Dyslipidaemia, an elevation in total cholesterol, or LDL-cholesterol, TAG or a reduction in HDL-cholesterol concentration in the circulation, is a potent risk factor for CVD(Reference Ballantyne, Arroll and Shepherd145). Atherosclerosis, the presence of extensive plaque (lipids, cholesterol, Ca and other substances) in the arteries, causes nearly 75 % of all deaths from CVD in the USA. Low HDL-cholesterol and high TAG concentrations are independent predictors of CVD(Reference Kendrick, Targher and Smits146). Elevated plasma lipoprotein(a) is an independent risk factor for the development of CHD in men, comparable in magnitude to a total cholesterol concentration ≥ 6·2 mmol/l or HDL-cholesterol < 0·9 mmol/l(Reference Bostom, Cupples and Jenner147). Using data from the National Heart, Lung, Blood Institute Family Heart Study, concentrations of lipoprotein(a)>50 mg/dl (500 mg/l) were shown to increase the risk of early onset of coronary artery disease, especially if combined with other unfavourable lipid levels or non-lipid risk factors, such as hypertension, diabetes or smoking(Reference Hopkins, Hunt and Schreiner148). Vitamin D status has been associated with dyslipidaemia in some instances but the literature is conflicting and the evidence basis is still being gathered.

Potential mechanisms linking vitamin D and lipids have not been studied in great detail and the focus has been on the effect of Ca, which implicates vitamin D indirectly through its role in maintaining Ca homeostasis. Increased Ca absorption reduces fatty acid absorption and increases faecal fatty acid content, most probably resulting from the formation of insoluble Ca–fatty soaps(Reference Barger-Lux, Heaney and Lanspa149Reference Reid151). The decreased absorption of fat, especially saturated fat, reduces serum total cholesterol and LDL-cholesterol. Furthermore, Ca increases the conversion of cholesterol to bile acids for excretion(Reference Vaskonen, Mervaala and Sumuvuori152) and increased Ca absorption can decrease hepatic TAG formation(Reference Cho, Kang and Choi153). An alternative mechanism may be a consequence of the suppressive effect of 1,25(OH)2D on PTH concentration. Raised PTH has been shown to be accompanied by a decrease in plasma post-heparin lipolytic activity(Reference Lacour, Basile and Drueke154); thus the maintenance of appropriate PTH levels with optimal vitamin D status may decrease TAG concentration by increasing peripheral TAG removal.

Clinical evidence – observational studies

Vitamin D status has been inversely related to dyslipidaemia in some instances, but the literature is conflicting. Lipid levels vary seasonally and total cholesterol, LDL-cholesterol and TAG concentrations are higher during the winter(Reference Kelly155). Many(Reference Liu, Meigs and Pittas80, Reference Martins, Wolf and Pan136, Reference Cigolini, Iagulli and Miconi156), but not all(Reference Chiu, Chu and Go53, Reference Auwerx, Bouillon and Kesteloot157, Reference Carbone, Rosenberg and Tolley158), studies have shown inverse relationships between 25(OH)D and TAG concentrations. Martins et al. (Reference Martins, Wolf and Pan136) showed that in US adults, TAG was lower in those in the top quartile of 25(OH)D ( ≥ 92·5 nmol/l) than in those in the bottom quartile ( < 52·5 nmol/l); however, there was no association between s25(OH)D and total cholesterol. In a smaller sample of 126 glucose-tolerant healthy adults, Chiu et al. (Reference Chiu, Chu and Go53) observed an inverse association between 25(OH)D and total cholesterol, and LDL-cholesterol (adjusting for sex, age, ethnicity, season, SBP and DBP, BMI, and waist-to-hip ratio); however, no association was apparent between 25(OH)D and HDL-cholesterol or TAG in this study. The sometimes reported association between s25(OH)D and HDL-cholesterol(Reference Liu, Meigs and Pittas80, Reference Auwerx, Bouillon and Kesteloot157, Reference Carbone, Rosenberg and Tolley158) is not apparent in all studies(Reference Chiu, Chu and Go53), but an association between 25(OH)D and apoA1 has been observed(Reference Auwerx, Bouillon and Kesteloot157Reference John, Noonan and Mannan159).

The total cholesterol:HDL and LDL:HDL ratios are important predictors of increased risk of CVD. Using data from the Framingham Offspring Study (n 1739), Wang et al. (Reference Wang, Pencina and Booth160) observed that the total cholesterol:HDL ratio was higher in those with s25(OH)D below 37·5 nmol/l. Although we saw no association between vitamin D status and lipid profiles in young European adults (S Muldowney, A Lucey, G Paschos, et al., unpublished results), we did observe associations in a representative sample of 12- and 15-year-old adolescents (M Kiely, S Muldowney, TR Hill, et al., unpublished results); those with a s25(OH)D below about 50 nmol/l were more likely to have elevated total cholesterol (OR 3·09; 95 % CI 1·33, 7·21) and LDL-cholesterol (OR 2·09; 95 % CI 0·95, 4·59) compared with adolescents who had a s25(OH)D>75 nmol/l. These associations were stronger in boys than girls.

Intervention trials

There is a dearth of RCT on the effects of cholecalciferol on lipids (see Table 1). Zittermann et al. (Reference Zittermann, Frisch and Berthold111) showed that treatment with cholecalciferol, parallel with energy restriction ( − 700 kcal; − 2930 kJ) resulted in a more pronounced decrease in TAG concentration ( − 13·5 % compared with +3 %), and an increase in LDL-cholesterol than did energy restriction alone. These beneficial effects on TAG concentration were independent of body weight, fat mass and sex. However, supplementation for 1 year with either 10 or 20 μg cholecalciferol in healthy Pakistani immigrants living in Denmark had no effect on total cholesterol, HDL-cholesterol, LDL-cholesterol or VLDL-cholesterol, the LDL:HDL ratio, or TAG concentration(Reference Andersen, Brot and Mejborn161), although the doses were probably not sufficient to achieve a clinically meaningful effect on lipids. At similar doses of between 5 and 15 μg/d, we showed no effect of cholecalciferol supplementation during the winter on lipids (S Muldowney, TR Hill, AJ Lucey, et al., unpublished results) in adults aged 20–40 and>65 years. On the basis of these data, future interventions need to use doses of vitamin D to achieve targeted s25(OH)D levels, which may have a beneficial effect on lipids. Four other RCT which reported insulin sensitivity/release or blood pressure as the primary outcome reported no effects on lipids(Reference Nagpal, Pande and Bhartia83, Reference von Hurst, Stonehouse and Coad85, Reference Jorde and Figenschau86, Reference Scragg, Khaw and Murphy139).

All other RCT investigating effects of cholecalciferol on lipids also gave Ca(Reference Jorde, Sneve and Torjesen89, Reference Major, Alarie and Dore109, Reference Gannage-Yared, Azoury and Mansour162Reference Daly and Nowson164). Gannage-Yared et al. (Reference Gannage-Yared, Azoury and Mansour162) observed no effect of 1 g Ca plus 20 μg cholecalciferol on LDL-cholesterol, or HDL-cholesterol or TAG concentration in postmenopausal women (n 39); however, this was a short-term trial of 12 weeks with a small number of subjects. Using data from the Women's Health Initiative, Rajpathak et al. (Reference Rajpathak, Xue and Wassertheil-Smoller163) observed no significant effect of 1 g Ca plus 10 μg cholecalciferol on circulating lipids after 5 years. Major et al. (Reference Major, Alarie and Dore109) showed that 1200 mg Ca plus 10 μg cholecalciferol for 15 weeks lowered total cholesterol:HDL and LDL:HDL ratios and LDL-cholesterol concentrations compared with placebo. These effects were independent of fat mass and waist circumference for the total cholesterol:HDL and LDL:HDL ratio but not for LDL-cholesterol. A recent RCT in which 140 men>50 years were randomised to receive either milk (400 ml of 1 % fat fortified with 1 g Ca and 20 μg vitamin D3) or no milk for 2 years showed no effect on total cholesterol, HDL-cholesterol, or LDL-cholesterol, or TAG concentration(Reference Daly and Nowson164). Jorde et al. (Reference Jorde, Sneve and Torjesen89) showed that 500 mg Ca per d plus 500 or 1000 μg cholecalciferol per week had no effect on lipid profiles after 1 year in 334 overweight and obese subjects.

For many years, treatment for psoriasis has included climate therapy. Osmancevic et al. (Reference Osmancevic, Nilsen and Landin-Wilhelmsen165) showed that psoriasis symptoms improved in twenty patients aged 25–65 years with 15 d of climate therapy in Gran Canaria. The LDL:HDL ratio decreased from 2·4 to 1·9 mmol/l (P < 0·001), apoA1 and HDL-cholesterol concentration increased and apoB decreased during the sun exposure period. However, there was no effect on either total or LDL-cholesterol. In contrast, Carbone et al. (Reference Carbone, Rosenberg and Tolley158) showed in young healthy adults (n 49) that UVB radiation had no effect on any lipoproteins or apolipoproteins. However, in this study, subgroup analysis showed differential effects of UVB radiation on lipids and lipoproteins according to baseline s25(OH)D concentrations, providing additional evidence for using a modified study design which seeks to achieve specific 25(OH)D levels and accounts for baseline levels in the data analysis.

There is some evidence that vitamin D may have a negative effect on lipids, particularly HDL-cholesterol, which requires further exploration. Postmenopausal women (n 402) in the Kuopio Osteoporosis Study were randomised to receive one of four treatments: (a) sequential combination of 2 mg oestradiol valerate and 1 mg cyproterone acetate (hormone replacement therapy); (b) 7·5 μg cholecalciferol; (c) hormone replacement therapy and 7·5 μg cholecalciferol; (d) placebo(Reference Tuppurainen, Heikkinen and Penttilä166). Both the cholecalciferol and placebo groups received 500 mg calcium lactate. The hormone replacement therapy treatment reduced serum total cholesterol, most probably due to the decrease in LDL-cholesterol; however, cholecalciferol supplementation increased LDL-cholesterol by 6 % in the 1-year period. In the follow-up study, Heikkinen et al. (Reference Heikkinen, Tuppurainen and Niskanen167) observed that cholecalciferol supplementation over 3 years lowered HDL-cholesterol. However, as this reduction was also observed in the placebo group, it may not have been an effect of vitamin D. A recent study showed that VDR knock-out mice had higher total and HDL-cholesterol than wild-type mice(Reference Wang, Keisala and Solakivi168). The higher HDL-cholesterol concentration was only apparent in males(Reference Wang, Keisala and Solakivi168) and requires replication in further studies before being acknowledged as a real effect. If vitamin D has a negative effect on HDL-cholesterol it may be a consequence of suppressive effects of 1,25(OH)2D on apoA1 gene expression, possibly by altering co-activators and co-repressors(Reference Wehmeier, Beers and Haas169). Overall, the evidence for a beneficial or negative effect of vitamin D on lipids requires much further exploration. With regard to total cholesterol, LDL-cholesterol and TAG, the evidence leans towards a beneficial association whereas the data on HDL-cholesterol are conflicting.

Inflammation

Obesity induces a chronic inflammatory environment, which is implicated in metabolic dysfunction and CVD(Reference Kowalska, Straczkowski and Nikolajuk170Reference Libby, Ridker and Maseri172). Pro-inflammatory cytokines include IL-1, IL-6 and TNF-α(Reference Stentz, Umpierrez and Cuervo173), whereas IL-10 has an anti-inflammatory effect(Reference Moore, de Waal Malefyt and Coffman174). C-reactive protein (CRP), an acute-phase reactant, is a clinically validated and widely measured marker of systemic inflammation and increased synthesis can be stimulated by IL-6 and TNF-α. Certain inflammatory cytokines and CRP levels are elevated in adults with obesity, CVD, type 2 diabetes and the MetS(Reference Kowalska, Straczkowski and Nikolajuk170, Reference Libby171, Reference Festa, D'Agostino and Howard175, Reference Ridker, Buring and Cook176). Accumulating evidence for an important role for vitamin D in regulation both of the adaptive and innate immune systems has led to intense interest in this field. Potent immunoregulatory effects of 1,25(OH)2D include anti-proliferative properties, stimulation of production of cathelicidin by macrophages and bacterial killing and modulation of B- and T-lymphocyte proliferation and function(Reference Adams and Hewison177). Most exciting about the recent discoveries is the role for 25(OH)D in induction of these effects, which means that the potential impact on human health by improving host defence and modulating the inflammatory response is immense(Reference Cantorna, Zhu and Froicu178).

Immune cells not only express the VDR but also CYP27B1(Reference Herder, Baumert and Thorand179, Reference Deluca and Cantorna180), allowing them to synthesise and secrete 1,25(OH)2D in a regulated fashion(Reference Mathieu and Adorini181). Crucially, as this occurs in an autocrine fashion and not under endocrine control, vitamin D-inactivating hydroxylase is also expressed, which helps to temper the local effects of 1,25(OH)2D, preventing potential damage caused by over-stimulation of the immune system(Reference Adams and Hewison177, Reference Overbergh, Decallonne and Valckx182). 1,25(OH)2D uses several different molecular mechanisms to regulate cytokine expression, either directly by targeting transcription initiation and regulation or indirectly interfering with other intracellular signalling pathways(Reference van Etten and Mathieu183). The 1,25(OH)2D/VDR/RXR complex can occupy the nuclear factor of activated T cells at the nuclear factor of activated T cells-1 site, in the promoter region of some cytokines, thereby preventing T cell-specific transcription factor forming a complex with activator protein 1, which is necessary for cytokine production(Reference van Etten, Stoffels and Gysemans184). Zhu et al. (Reference Zhu, Mahon and Froicu185) showed that in colonic tissue of IL-10 knock-out mice with inflammatory bowel disease, 1,25(OH)2D was capable of down-regulating several genes associated with TNF-α, including TNF-α receptor 1, proteins involved in the transcription of TNF-α, and TNF-α itself. As well as direct effects, vitamin D could exert beneficial effects on cytokine production indirectly by maintaining serum Ca concentrations; in human umbilical vein cord endothelial cells, hypocalcaemia up-regulated endothelial NF-κB activity and IL-6 concentration was elevated(Reference Talmor-Barkan, Rashid and Weintal186). In addition, PTH induces IL-6 synthesis in osteoblasts(Reference Greenfield, Gornik and Horowitz187).

Clinical data

Data examining the associations between vitamin D and inflammation in relation to CVD are limited and the data are conflicting. In type 2 diabetics (n 459), CRP concentration was higher in those with a s25(OH)D < 50 compared with those above 50 nmol/l(Reference Cigolini, Iagulli and Miconi156). A cross-sectional analysis of the Framingham Offspring Study (n 1381; aged 35–89 years) showed that plasma 25(OH)D was not associated with overall inflammation, indicated by the inflammation index, sum of the fourteen individual markers of inflammation used in the study(Reference Shea, Booth and Massaro188). In addition, plasma 25(OH)D was not associated with any of the individual markers except IL-6 (P = 0·02). Peterson & Heffernan(Reference Peterson and Heffernan189) showed in healthy women (n 69), aged 25–82 years, that s25(OH)D concentration was inversely associated with TNF-α but no association was apparent between s25(OH)D and IL-6, IL-10 or CRP. Unexpectedly, we showed in young overweight adults that having a s25(OH)D concentration < 50 nmol/l decreased the likelihood (OR 0·35; 95 % CI 0·17, 0·73) of having an elevated CRP concentration(S Muldowney, A Lucey, G Paschos, et al., unpublished results).

Intervention trials

Data from RCT looking at the effects of vitamin D3 (as a single treatment) on inflammatory cytokines or CRP in healthy subjects are meagre. Most of the evidence comes from studies where participants have underlying conditions, or vitamin D deficiency (see Table 1). A total of twenty-two intensive care unit patients were randomised to receive daily either 5 μg (low dose) or 12·5 μg (high dose) for 10 d and compared with healthy age-, sex- and BMI-matched controls(Reference Van den Berghe, Van Roosbroeck and Vanhove190). While treatment had no effect on TNF-α or IL-1, the reduction in CRP was greater in the high-dose group compared with the low-dose group, and a reduction in IL-6 was only apparent in the high-dose group. Timms et al. (Reference Timms, Mannan and Hitman71) randomised healthy British adults of Bangladeshi origin, with vitamin D deficiency (s25(OH)D < 27·5 nmol/l), to receive 3-monthly injections of an oily cholecalciferol solution (1250 μg or 12·5 μg) for 1 year. The two treatment groups were combined and CRP concentration was dramatically reduced post-supplementation ( − 23 %). In the Zittermann et al. (Reference Zittermann, Frisch and Berthold111) weight-loss study, compared with placebo, vitamin D supplementation decreased TNF-α concentration, whereas there was no effect on CRP or IL-6 concentration in either group. Most recently, eighty-one South Asian women living in New Zealand, who were insulin resistant and had a s25(OH)D < 50 nmol/l, were randomised to receive either 100 μg cholecalciferol or a placebo daily for 6 months; however, no effect on CRP was apparent(Reference von Hurst, Stonehouse and Coad85). Similarly, we observed no effect on CRP after 6 months of cholecalciferol supplementation in healthy adults (S Muldowney, TR Hill, AJ Lucey, et al., unpublished results).

To the best of our knowledge all other vitamin D RCT looking at its effects on inflammatory markers were in combination with Ca. Schleithoff et al. (Reference Schleithoff, Zittermann and Tenderich191) supplemented congestive heart failure patients (n 93) with 500 mg Ca combined with either 50 μg cholecalciferol daily or a placebo for 9 months. In patients who received vitamin D a reduction in TNF-α concentration was apparent; in addition, IL-10 concentration was increased. Multiple sclerosis patients (n 39) with a s25(OH)D concentration < 50 nmol/l were randomised to receive daily 800 mg Ca plus placebo or 800 mg Ca plus 25 μg cholecalciferol for 6 months(Reference Mahon, Gordon and Cruz192). Vitamin D supplementation increased serum concentrations of transforming growth factor-β1, an anti-inflammatory cytokine, whereas there was no effect in the placebo group. However, mRNA levels of TNF-α, IL-2, interferon-γ and IL-13 concentrations did not change in either group. In a double-blind randomised study, where non-diabetic elderly adults (aged>65 years) were given either 500 mg Ca and 17·5 μg cholecalciferol or a placebo, no effect on CRP or IL-6 concentration was observed over 3 years in either subjects with impaired fasting glucose or normal fasting glucose concentrations(Reference Pittas, Harris and Stark87). A short-term supplementation trial (12-week duration) with 1000 mg Ca and 20 μg cholecalciferol daily of postmenopausal women (n 39) had no effect on circulating concentrations of IL-6, TNF-α or CRP(Reference Andersen, Brot and Mejborn161). More recently, a large RCT on overweight and obese subjects (n 334) showed no effect of supplementation with Ca (500 mg/d) plus cholecalciferol (500 or 1000 μg/week) for 1 year on a wide range of cytokines and other inflammatory markers(Reference Jorde, Sneve and Torjesen193).

In brief, there appears to be no effect of supplementation in healthy adults (S Muldowney, TR Hill, AJ Lucey, et al., unpublished results; Pittas et al. (Reference Pittas, Harris and Stark87); Jorde et al. (Reference Jorde, Sneve and Torjesen89); Gannage-Yared et al. (Reference Gannage-Yared, Azoury and Mansour162)); unless vitamin D deficient at baseline(Reference Timms, Mannan and Hitman71), this may be related to the low circulating concentration of the cytokines in healthy subjects. Furthermore, the studies by Schleithoff et al. (Reference Schleithoff, Zittermann and Tenderich191) and Mahon et al. (Reference Mahon, Gordon and Cruz192) suggest that a high requirement of vitamin D (>25 μg/d) is needed to mediate its immunological protective effects.

Novel markers of CVD risk

Adhesion molecules, PAI-1 and matrix metalloproteinases (MMP) and their inhibitors can be used as markers of CVD risk and, recently, have been associated with vitamin D status. The adhesion molecules, such as intracellular- and vascular-cell adhesion molecules, mediate the migration of inflammatory cells to atherosclerotic lesions. They act as ligands to tightly bind various molecules in the inflammatory process. A small number of studies, with conflicting results, have shown a relationship between vitamin D (1,25(OH)2D) and the adhesion molecules(Reference Weinreich, Wuthrich and Booy194Reference Kreft, Brzoska and Doehn197). The positive effect is most probably mediated through 1,25(OH)2D action on suppressing TNF-α secretion(Reference Van den Berghe, Van Roosbroeck and Vanhove190), but further studies are required to elucidate this relationship further.

PAI-1, which is produced by the vascular endothelium and platelets, is an important regulator of fibrinolysis(Reference Kohler and Grant198) as it rapidly binds, and therefore inhibits, tissue plasminogen activator and urinary plasminogen activator. High PAI-1 concentrations have been shown to predict first occurrence of a myocardial infarction(Reference Thogersen, Jansson and Boman199), or reoccurrence before the age of 45 years(Reference Hamsten, Wiman and de Faire200, Reference Hamsten, de Faire and Walldius201). Studies have found that 1,25(OH)2D reduces PAI-1 concentrations in various cell types, including vascular cells and smooth-muscle cells(Reference Wu-Wong, Nakane and Ma202, Reference Wu-Wong, Nakane and Ma203). A possible mechanism for the association between PAI-1 and vitamin D may be similar to blood pressure, which involves the RAS. Angiotensin II stimulates the production of PAI-1 in cultured endothelial and vascular smooth-muscle cells(Reference Vaughan, Lazos and Tong204), whereas the inhibition of angiotensin-converting enzyme is associated with a decrease in both plasma PAI-1 concentrations and PAI-1 activity(Reference Brown, Agirbasli and Vaughan205). Recently, a large RCT with Ca (500 mg daily plus cholecalciferol at 500 or 1000 μg/week) for 1 year showed no effect on PAI-1 or other haemostatic markers in overweight and obese subjects(Reference Jorde, Sneve and Torjesen206). However, evidence is limited and more studies (cross-sectional, interventional and mechanistic) are required.

MMP are a family of Zn-containing enzymes that have an important role in extracellular matrix degradation, synthesis and remodelling(Reference Rahman, Hershey and Ahmed207). MMP are activated by MMP proenzymes and inhibited by endogenous tissue inhibitors of metalloproteinases (TIMP) and the net proteolytic activity is a function of the balance of MMP to TIMP. MMP degradation of the extracellular matrix may assist the influx of leucocytes through the endothelial layer, contributing to a decrease in endothelial barrier function, and facilitate migration of vascular smooth muscle cells into the intimal space where they proliferate and contribute to plaque formation(Reference Libby, Ridker and Maseri172, Reference Dollery and Libby208). MMP-9 and TIMP-1 have been identified as novel predictors of CVD risk. Experimental studies (either animal or cell) have shown that 1,25(OH)2D regulates MMP expression(Reference Li, Kong and Wei118, Reference Rahman, Hershey and Ahmed207, Reference Dean, Schwartz and Schmitz209). Rahman et al. (Reference Rahman, Hershey and Ahmed207) showed that TIMP-1 and TIMP-3 were significantly under-expressed, while MMP-2 and MMP-9 were up-regulated in the cardiac tissue of VDR knock-out mice as compared with wild-type mice. However, human studies supporting an association between vitamin D and the MMP/TIMP system in humans are limited. A cross-sectional study, ran in tangent with an intervention study on healthy Bangladeshi adults living in Britain (n 171; aged 35–65 years), who were vitamin D deficient (s25(OH)D < 27·5 nmol/l) showed that vitamin D status was an independent predictor of MMP-9 concentration and a reduction in MMP-9 (66·8 %) and TIMP-1 (39·8 %) concentration was apparent following vitamin D supplementation(Reference Timms, Mannan and Hitman71).

Summary and future directions

There is a considerable body of evidence from cross-sectional and prospective studies showing that low vitamin D status is associated with an increased risk of CVD, the MetS and each of its individual risk factors. Furthermore, there are strong mechanistic data to support the role of vitamin D in the reduction of CVD risk factors, particularly for insulin secretion, insulin resistance and hypertension mediated by the RAS. Evidence is not as robust for the association between vitamin D status and dyslipidaemia and further studies are required. Data from RCT are relatively rare and are confounded by inappropriate study design, inadequate characterisation of subjects, lack of data on season and sunshine exposure and interventions combining Ca and vitamin D.

If vitamin D influences cardiometabolic health, these effects may only be measurable at higher doses than those achievable through diet. This raises several questions about the food/pharma interface with respect to nutrition interventions, the impact that high dose studies have on skewing the literature and how such data should be considered when dietary recommendations are under review. With the notable exception of folic acid, where supplemental approaches are required to achieve the maximum benefit, nutritional science so far has relied on food-based approaches to achieve optimal health through diet. However, increasing confidence about the safety of vitamin D doses in excess of the current tolerable upper intake levels should enable further RCT to be implemented using vitamin D as a single treatment at doses calibrated to achieve target thresholds of s25(OH)D concentrations, particularly in vitamin D-deficient individuals or patient subgroups with elevated CVD risk biomarkers.

Acknowledgements

The present review received no specific grant from any funding agency in the public, commercial or not-for-profit sectors.

Three studies submitted for publication have been included as unpublished results. These are: Muldowney S, Lucey A, Paschos G, et al. ‘Vitamin D status and cardio-metabolic risk factors in young European adults’; Kiely M, Muldowney S, Hill TR, et al. ‘Vitamin D status and cardio-metabolic risk factors in adolescents’; Muldowney S, Hill TR, Lucey AJ, et al. ‘Effect of vitamin D supplementation on biomarkers of CVD risk in 20–40 and 64+ year old men and women’.

Both authors contributed equally to the drafting and editing of the manuscript.

The authors have no conflict of interest.

References

1Lloyd-Jones, D, Adams, R, Carnethon, M, et al. . (2009) Heart Disease and Stroke Statistics – 2009 Update: A Report From the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Circulation 119, e21e181.Google ScholarPubMed
2Central Statistics Office (2007) Deaths from Principal Causes Registered in the Years 1998 to 2005. Cork, Republic of Ireland: Central Statistics Office.Google Scholar
3Thom, T, Haase, N, Rosamond, W, et al. . (2006) Heart disease and stroke statistics – 2006 update: a report from the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Circulation 113, e85e151.Google ScholarPubMed
4Yusuf, SH, Hawken, S, Ounpuu, S, et al. . (2004) Effect of potentially modifiable risk factors associated with myocardial infarction in 52 countries (the INTERHEART study): case–control study. Lancet 364, 937952.CrossRefGoogle ScholarPubMed
5Kylin, E (1923) Studien uber das Hypertonie-Hyperglyka ‘mie-Hyperurika’ Miesyndrom (Studies of the hypertension–hyperglycemia–hyperuricemia syndrome). Zentralbl Inn Med 44, 105127.Google Scholar
6Ford, ES, Giles, WH & Dietz, WH (2002) Prevalence of the metabolic syndrome among US adults: findings from the third National Health and Nutrition Examination Survey. JAMA 287, 356359.CrossRefGoogle ScholarPubMed
7Hu, G, Qiao, Q, Tuomilehto, J, et al. . (2004) Prevalence of the metabolic syndrome and its relation to all-cause and cardiovascular mortality in nondiabetic European men and women. Arch Intern Med 164, 10661076.Google Scholar
8Villegas, RP, Perry, IJ, Creagh, D, et al. . (2003) Prevalence of the metabolic syndrome in middle-aged men and women. Diabetes Care 26, 31983199.Google Scholar
9World Health Organization (1999) Definition, Diagnosis and Classification of Diabetes Mellitus and its Complications. Geneva: Department of Noncommunicable Disease Surveillance.Google Scholar
10National Institutes of Health (2001) Third Report of the National Cholesterol Education Program Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III): Executive Summary. Bethesda, MD: National Institutes of Health.Google Scholar
11Ford, ES, Ajani, UA, McGuire, LC, et al. . (2005) Concentrations of serum vitamin D and the metabolic syndrome among U.S. adults. Diabetes Care 28, 12281230.CrossRefGoogle ScholarPubMed
12Dobnig, H, Pilz, S, Scharnagl, H, et al. . (2008) Independent association of low serum 25-hydroxyvitamin D and 1,25-dihydroxyvitamin D levels with all-cause and cardiovascular mortality. Arch Intern Med 168, 13401349.CrossRefGoogle Scholar
13Holick, MF, Biancuzzo, RM, Chen, TC, et al. . (2008) Vitamin D2 is as effective as vitamin D3 in maintaining circulating concentrations of 25-hydroxyvitamin D. J Clin Endocrinol Metab 93, 677681.CrossRefGoogle Scholar
14Abdelkefi, A, Achour, W, Ben Othman, T, et al. . (2007) Use of heparin-coated central venous lines to prevent catheter-related bloodstream infection. J Support Oncol 5, 273278.Google ScholarPubMed
15Holick, MF (2004) Vitamin D: importance in the prevention of cancers, type 1 diabetes, heart disease, and osteoporosis. Am J Clin Nutr 79, 362371.CrossRefGoogle ScholarPubMed
16Haussler, MR, Whitfield, GK, Haussler, CA, et al. . (1998) The nuclear vitamin D receptor: biological and molecular regulatory properties revealed. J Bone Miner Res 13, 325349.CrossRefGoogle ScholarPubMed
17Zittermann, A (2003) Vitamin D in preventive medicine: are we ignoring the evidence? Br J Nutr 89, 552572.CrossRefGoogle ScholarPubMed
18Hass, GM, Landerholm, W & Hemmens, A (1966) Production of calcific athero-arteriosclerosis and thromboarteritis with nicotine, vitamin D and dietary cholesterol. Am J Pathol 49, 739771.Google Scholar
19Eisenstein, R, Ellis, H & Rosato, J (1969) In vitro studies of vitamin D-induced aortic calcification. Proc Soc Exp Biol Med 132, 5862.CrossRefGoogle ScholarPubMed
20Morrison, LM, Bajwa, GS, Alfin-Slater, RB, et al. . (1972) Prevention of vascular lesions by chondroitin sulfate A in the coronary artery and aorta of rats induced by a hypervitaminosis D, cholesterol-containing diet. Atherosclerosis 16, 105118.CrossRefGoogle ScholarPubMed
21Linden, V (1974) Vitamin D and myocardial infarction. Br Med J 3, 647650.Google Scholar
22Schmidt-Gayk, H, Goossen, J, Lendle, F, et al. . (1977) Serum 25-hydroxycalciferol in myocardial infarction. Atherosclerosis 26, 5558.CrossRefGoogle ScholarPubMed
23Vik, B, Try, K, Thelle, DS, et al. . (1979) Tromso Heart Study: vitamin D metabolism and myocardial infarction. Br Med J 2, 176.Google Scholar
24Zittermann, A & Koerfer, R (2008) Vitamin D in the prevention and treatment of coronary heart disease. Curr Opin Clin Nutr Metab Care 11, 752757.Google Scholar
25Zittermann, A, Schleithoff, SS & Koerfer, R (2005) Putting cardiovascular disease and vitamin D insufficiency into perspective. Br J Nutr 94, 483492.CrossRefGoogle ScholarPubMed
26Grimes, DS, Hindle, E & Dyer, T (1996) Sunlight, cholesterol and coronary heart disease. Q J Med 89, 579590.Google Scholar
27Giovannucci, E, Liu, Y, Hollis, BW, et al. . (2008) 25-Hydroxyvitamin D and risk of myocardial infarction in men: a prospective study. Arch Intern Med 168, 11741180.CrossRefGoogle Scholar
28Trivedi, DP, Doll, R & Khaw, KT (2003) Effect of four monthly oral vitamin D3 (cholecalciferol) supplementation on fractures and mortality in men and women living in the community: randomised double blind controlled trial. BMJ 326, 469.CrossRefGoogle ScholarPubMed
29Hsia, J, Heiss, G, Ren, H, et al. . (2007) Calcium/vitamin D supplementation and cardiovascular events. Circulation 115, 846854.CrossRefGoogle ScholarPubMed
30LaCroix, AZ, Kotchen, J, Anderson, G, et al. . (2009) Calcium plus vitamin D supplementation and mortality in postmenopausal women: the Women's Health Initiative calcium–vitamin D randomized controlled trial. J Gerontol A Biol Sci Med Sci 64, 559567.CrossRefGoogle ScholarPubMed
31Prince, RL, Austin, N, Devine, A, et al. . (2008) Effects of ergocalciferol added to calcium on the risk of falls in elderly high-risk women. Arch Intern Med 168, 103108.CrossRefGoogle ScholarPubMed
32Bolland, MJ, Avenell, A, Baron, JA, et al. . (2010) Effect of calcium supplements on risk of myocardial infarction and cardiovascular events: meta analysis. BMJ 341, c3691.Google Scholar
33Alberti, KG, Zimmet, P & Shaw, J (2005) The metabolic syndrome – a new worldwide definition. Lancet 366, 10591062.Google Scholar
34Einhorn, D, Reaven, GM, Cobin, RH, et al. . (2003) American College of Endocrinology position statement on the insulin resistance syndrome. Endocr Pract 9, 237252.Google Scholar
35Grundy, SM, Cleeman, JI, Daniels, SR, et al. . (2005) Diagnosis and management of the metabolic syndrome. An American Heart Association/National Heart, Lung, and Blood Institute Scientific Statement. Executive summary. Cardiol Rev 13, 322327.CrossRefGoogle ScholarPubMed
36Isomaa, B, Almgren, P, Tuomi, T, et al. (2001) Cardiovascular morbidity and mortality associated with the metabolic syndrome. Diabetes Care 24, 683689.Google Scholar
37Forouhi, NG, Luan, J, Cooper, A, et al. . (2008) 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 57, 26192625.Google Scholar
38Hypponen, E, Boucher, BJ, Berry, DJ, et al. . (2008) 25-Hydroxyvitamin D, IGF-1, and metabolic syndrome at 45 years of age: a cross-sectional study in the 1958 British Birth Cohort. Diabetes 57, 298305.Google Scholar
39Reis, JP, von Muhlen, D & Miller, ER III (2008) Relation of 25-hydroxyvitamin D and parathyroid hormone levels with metabolic syndrome among US adults. Eur J Endocrinol 159, 4148.Google Scholar
40Hjelmesaeth, J, Hofso, D, Aasheim, ET, et al. . (2009) Parathyroid hormone, but not vitamin D, is associated with the metabolic syndrome in morbidly obese women and men: a cross-sectional study. Cardiovasc Diabetol 8, 7.CrossRefGoogle Scholar
41McGill, AT, Stewart, JM, Lithander, FE, et al. . (2008) Relationships of low serum vitamin D3 with anthropometry and markers of the metabolic syndrome and diabetes in overweight and obesity. Nutr J 7, 4.CrossRefGoogle ScholarPubMed
42Reis, JP, von Muhlen, D, Kritz-Silverstein, D, et al. . (2007) Vitamin D, parathyroid hormone levels, and the prevalence of metabolic syndrome in community-dwelling older adults. Diabetes Care 30, 15491555.Google Scholar
43Parker, J, Hashmi, O, Dutton, D, et al. . (2010) Levels of vitamin D and cardiometabolic disorders: systematic review and meta-analysis. Maturitas 65, 225236.CrossRefGoogle ScholarPubMed
44Weyer, C, Bogardus, C & Pratley, RE (1999) Metabolic factors contributing to increased resting metabolic rate and decreased insulin-induced thermogenesis during the development of type 2 diabetes. Diabetes 48, 16071614.CrossRefGoogle ScholarPubMed
45Pittas, AG, Lau, J, Hu, FB, et al. . (2007) The role of vitamin D and calcium in type 2 diabetes. A systematic review and meta-analysis. J Clin Endocrinol Metab 92, 20172029.Google Scholar
46Teegarden, D & Donkin, SS (2009) Vitamin D: emerging new roles in insulin sensitivity. Nutr Res Rev 22, 8292.CrossRefGoogle ScholarPubMed
47Bland, R, Markovic, D, Hills, CE, et al. . (2004) Expression of 25-hydroxyvitamin D3-1α-hydroxylase in pancreatic islets. J Steroid Biochem Mol Biol 89–90, 121125.CrossRefGoogle ScholarPubMed
48Zeitz, U, Weber, K, Soegiarto, DW, et al. . (2003) Impaired insulin secretory capacity in mice lacking a functional vitamin D receptor. FASEB J 17, 509511.Google Scholar
49Bourlon, PM, Billaudel, B & Faure-Dussert, A (1999) Influence of vitamin D3 deficiency and 1,25 dihydroxyvitamin D3 on de novo insulin biosynthesis in the islets of the rat endocrine pancreas. J Endocrinol 160, 8795.Google Scholar
50Arnold, BM, Kuttner, M, Swaminathan, R, et al. . (1975) Radioimmunoassay studies of intestinal calcium-binding protein in the pig. I. Identification of intestinal calcium-binding protein in blood and response to a low calcium diet. Can J Physiol Pharmacol 53, 11291134.Google Scholar
51Morrissey, RL, Bucci, TJ, Richard, B, et al. . (1975) Calcium-binding protein: its cellular localization in jejunum, kidney and pancreas. Proc Soc Exp Biol Med 149, 5660.CrossRefGoogle ScholarPubMed
52Sooy, K, Schermerhorn, T, Noda, M, et al. . (1999) Calbindin-D28k controls [Ca(2+)]i and insulin release. Evidence obtained from calbindin-D28k knockout mice and β cell lines. J Biol Chem 274, 3434334349.Google Scholar
53Chiu, KC, Chu, A, Go, VL, et al. . (2004) Hypovitaminosis D is associated with insulin resistance and β cell dysfunction. Am J Clin Nutr 79, 820825.CrossRefGoogle ScholarPubMed
54Calle, C, Maestro, B & Garcia-Arencibia, M (2008) Genomic actions of 1,25-dihydroxyvitamin D3 on insulin receptor gene expression, insulin receptor number and insulin activity in the kidney, liver and adipose tissue of streptozotocin-induced diabetic rats. BMC Mol Biol 9, 65.Google Scholar
55Chiu, KC, Chuang, LM, Lee, NP, et al. . (2000) Insulin sensitivity is inversely correlated with plasma intact parathyroid hormone level. Metabolism 49, 15011505.Google Scholar
56McCarty, MF & Thomas, CA (2003) PTH excess may promote weight gain by impeding catecholamine-induced lipolysis – implications for the impact of calcium, vitamin D, and alcohol on body weight. Med Hypotheses 61, 535542.Google Scholar
57Wareham, NJ, Byrne, CD, Carr, C, et al. . (1997) Glucose intolerance is associated with altered calcium homeostasis: a possible link between increased serum calcium concentration and cardiovascular disease mortality. Metabolism 46, 11711177.Google Scholar
58Snijder, M, van Dam, R, Visser, M, et al. . (2006) To: Mathieu C, Gysemans C, Giulietti A, Bouillon R (2005) Vitamin D and diabetes. Diabetologia 48, 1247–1257. Diabetologia 49, 217218.CrossRefGoogle Scholar
59Reis, AF, Hauache, OM & Velho, G (2005) Vitamin D endocrine system and the genetic susceptibility to diabetes, obesity and vascular disease. A review of evidence. Diabetes Metab 31, 318325.CrossRefGoogle ScholarPubMed
60Palomer, X, Gonzalez-Clemente, JM, Blanco-Vaca, F, et al. . (2008) Role of vitamin D in the pathogenesis of type 2 diabetes mellitus. Diabetes Obes Metab 10, 185197.Google Scholar
61Dilmec, F, Uzer, E, Akkafa, F, et al. . (2010) Detection of VDR gene ApaI and TaqI polymorphisms in patients with type 2 diabetes mellitus using PCR-RFLP method in a Turkish population. J Diabetes Complications 24, 186191.Google Scholar
62Ye, WZ, Dubois-Laforgue, D, Bellanne-Chantelot, C, et al. . (2001) Variations in the vitamin D-binding protein (Gc locus) and risk of type 2 diabetes mellitus in French Caucasians. Metabolism 50, 366369.Google Scholar
63Malecki, MT, Frey, J, Moczulski, D, et al. . (2003) Vitamin D receptor gene polymorphisms and association with type 2 diabetes mellitus in a Polish population. Exp Clin Endocrinol Diabetes 111, 505509.CrossRefGoogle Scholar
64Hitman, GA, Mannan, N, McDermott, MF, et al. . (1998) Vitamin D receptor gene polymorphisms influence insulin secretion in Bangladeshi Asians. Diabetes 47, 688690.Google Scholar
65Ogunkolade, BW, Boucher, BJ, Prahl, JM, et al. . (2002) Vitamin D receptor (VDR) mRNA and VDR protein levels in relation to vitamin D status, insulin secretory capacity, and VDR genotype in Bangladeshi Asians. Diabetes 51, 22942300.Google Scholar
66Oh, JY & Barrett-Connor, E (2002) Association between vitamin D receptor polymorphism and type 2 diabetes or metabolic syndrome in community-dwelling older adults: the Rancho Bernardo Study. Metabolism 51, 356359.Google Scholar
67Tworowska-Bardzinska, U, Lwow, F, Kubicka, E, et al. . (2008) The vitamin D receptor gene BsmI polymorphism is not associated with anthropometric and biochemical parameters describing metabolic syndrome in postmenopausal women. Gynecol Endocrinol 24, 514518.CrossRefGoogle Scholar
68Chiu, KC, Chuang, LM & Yoon, C (2001) The vitamin D receptor polymorphism in the translation initiation codon is a risk factor for insulin resistance in glucose tolerant Caucasians. BMC Med Genet 2, 2.CrossRefGoogle ScholarPubMed
69Filus, A, Trzmiel, A, Kuliczkowska-Plaksej, J, et al. . (2008) Relationship between vitamin D receptor BsmI and FokI polymorphisms and anthropometric and biochemical parameters describing metabolic syndrome. Aging Male 11, 134139.Google Scholar
70Tai, K, Need, AG, Horowitz, M, et al. . (2008) Vitamin D, glucose, insulin, and insulin sensitivity. Nutrition 24, 279285.Google Scholar
71Timms, PM, Mannan, N, Hitman, GA, et al. . (2002) Circulating MMP9, vitamin D and variation in the TIMP-1 response with VDR genotype: mechanisms for inflammatory damage in chronic disorders? QJM 95, 787796.CrossRefGoogle ScholarPubMed
72Duncan, BB, Schmidt, MI, Pankow, JS, et al. . (2003) Low-grade systemic inflammation and the development of type 2 diabetes: the atherosclerosis risk in communities study. Diabetes 52, 17991805.CrossRefGoogle ScholarPubMed
73Pradhan, AD, Manson, JE, Rifai, N, et al. . (2001) C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. JAMA 286, 327334.Google Scholar
74Giulietti, A, van Etten, E, Overbergh, L, et al. . (2007) Monocytes from type 2 diabetic patients have a pro-inflammatory profile. 1,25-Dihydroxyvitamin D3 works as anti-inflammatory. Diabetes Res Clin Pract 77, 4757.Google Scholar
75Choi, HK, Willett, WC, Stampfer, MJ, et al. . (2005) Dairy consumption and risk of type 2 diabetes mellitus in men: a prospective study. Arch Intern Med 165, 9971003.CrossRefGoogle ScholarPubMed
76Liu, S, Song, Y, Ford, ES, et al. . (2005) Dietary calcium, vitamin D, and the prevalence of metabolic syndrome in middle-aged and older U.S. women. Diabetes Care 28, 29262932.CrossRefGoogle ScholarPubMed
77Liu, S, Choi, HK, Ford, E, et al. . (2006) A prospective study of dairy intake and the risk of type 2 diabetes in women. Diabetes Care 29, 15791584.Google Scholar
78Pittas, AG, Dawson-Hughes, B, Li, T, et al. . (2006) Vitamin D and calcium intake in relation to type 2 diabetes in women. Diabetes Care 29, 650656.CrossRefGoogle ScholarPubMed
79Scragg, R, Sowers, M & Bell, C (2004) Serum 25-hydroxyvitamin D, diabetes, and ethnicity in the Third National Health and Nutrition Examination Survey. Diabetes Care 27, 28132818.Google Scholar
80Liu, E, Meigs, JB, Pittas, AG, et al. . (2009) Plasma 25-hydroxyvitamin D is associated with markers of the insulin resistant phenotype in nondiabetic adults. J Nutr 139, 329334.Google Scholar
81Scragg, R, Holdaway, I, Singh, V, et al. . (1995) Serum 25-hydroxyvitamin D3 levels decreased in impaired glucose tolerance and diabetes mellitus. Diabetes Res Clin Pract 27, 181188.CrossRefGoogle ScholarPubMed
82Hypponen, E & Power, C (2006) Vitamin D status and glucose homeostasis in the 1958 British birth cohort: the role of obesity. Diabetes Care 29, 22442246.Google Scholar
83Nagpal, J, Pande, JN & Bhartia, A (2009) A double-blind, randomized, placebo-controlled trial of the short-term effect of vitamin D3 supplementation on insulin sensitivity in apparently healthy, middle-aged, centrally obese men. Diabet Med 26, 1927.CrossRefGoogle ScholarPubMed
84Borissova, AM, Tankova, T, Kirilov, G, et al. . (2003) The effect of vitamin D3 on insulin secretion and peripheral insulin sensitivity in type 2 diabetic patients. Int J Clin Pract 57, 258261.Google Scholar
85von Hurst, PR, Stonehouse, W & Coad, J (2010) 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 103, 549555.Google Scholar
86Jorde, R & Figenschau, Y (2009) Supplementation with cholecalciferol does not improve glycaemic control in diabetic subjects with normal serum 25-hydroxyvitamin D levels. Eur J Nutr 48, 349354.Google Scholar
87Pittas, AG, Harris, SS, Stark, PC, et al. . (2007) The effects of calcium and vitamin D supplementation on blood glucose and markers of inflammation in nondiabetic adults. Diabetes Care 30, 980986.Google Scholar
88Dawson-Hughes, B, Harris, SS, Krall, EA, et al. . (1997) Effect of calcium and vitamin D supplementation on bone density in men and women 65 years of age or older. N Engl J Med 337, 670676.CrossRefGoogle ScholarPubMed
89Jorde, R, Sneve, M, Torjesen, P, et al. . (2010) No improvement in cardiovascular risk factors in overweight and obese subjects after supplementation with vitamin D3 for 1 year. J Intern Med 267, 462472.CrossRefGoogle ScholarPubMed
90de Boer, IH, Tinker, LF, Connelly, S, et al. . (2008) Calcium plus vitamin D supplementation and the risk of incident diabetes in the Women's Health Initiative. Diabetes Care 31, 701707.Google Scholar
91World Health Organization (2006) Obesity and Overweight. Fact sheet no. 311. http://www.who.int/mediacentre/factsheets/fs311/en/index.html (accessed November 2008).Google Scholar
92Flegal, KM, Carroll, MD, Ogden, CL, et al. . (2002) Prevalence and trends in obesity among US adults, 1999–2000. JAMA 288, 17231727.CrossRefGoogle ScholarPubMed
93McCarthy, SN, Harrington, KE, Kiely, M, et al. . (2001) Analyses of the anthropometric data from the North/South Ireland Food Consumption Survey. Public Health Nutr 4, 10991106.Google Scholar
94Lee, P & Cunningham, K (1990) The Irish National Nutrition Survey. Dublin: Irish Nutrition and Dietetic Institute.Google Scholar
95Pérez-López, F (2009) Vitamin D metabolism and cardiovascular risk factors in postmenopausal women. Maturitas 62, 248262.Google Scholar
96Jorde, R, Sneve, M, Emaus, N, et al. . (2010) Cross-sectional and longitudinal relation between serum 25-hydroxyvitamin D and body mass index: the Tromso study. Eur J Nutr 49, 401407.Google Scholar
97Compston, JE, Vedi, S, Ledger, JE, et al. . (1981) Vitamin D status and bone histomorphometry in gross obesity. Am J Clin Nutr 34, 23592363.Google Scholar
98Harris, SS & Dawson-Hughes, B (2007) Reduced sun exposure does not explain the inverse association of 25-hydroxyvitamin D with percent body fat in older adults. J Clin Endocrinol Metab 92, 31553157.Google Scholar
99Bell, NH, Epstein, S, Greene, A, et al. . (1985) Evidence for alteration of the vitamin D–endocrine system in obese subjects. J Clin Invest 76, 370373.Google Scholar
100Shi, H, Norman, AW, Okamura, WH, et al. . (2001) 1α,25-Dihydroxyvitamin D3 modulates human adipocyte metabolism via nongenomic action. FASEB J 15, 27512753.Google Scholar
101Zemel, MB, Shi, H, Greer, B, et al. . (2000) Regulation of adiposity by dietary calcium. FASEB J 14, 11321138.Google Scholar
102Parikh, SJ, Edelman, M, Uwaifo, GI, et al. . (2004) The relationship between obesity and serum 1,25-dihydroxy vitamin D concentrations in healthy adults. J Clin Endocrinol Metab 89, 11961199.Google Scholar
103Konradsen, S, Ag, H, Lindberg, F, et al. . (2008) Serum 1,25-dihydroxy vitamin D is inversely associated with body mass index. Eur J Nutr 47, 8791.CrossRefGoogle Scholar
104Wortsman, J, Matsuoka, LY, Chen, TC, et al. . (2000) Decreased bioavailability of vitamin D in obesity. Am J Clin Nutr 72, 690693.Google Scholar
105Arunabh, S, Pollack, S, Yeh, J, et al. . (2003) Body fat content and 25-hydroxyvitamin D levels in healthy women. J Clin Endocrinol Metab 88, 157161.CrossRefGoogle ScholarPubMed
106Freedman, BI, Wagenknecht, LE, Hairston, KG, et al. . (2010) Vitamin D, adiposity, and calcified atherosclerotic plaque in African-Americans. J Clin Endocrinol Metab 95, 10761083.CrossRefGoogle ScholarPubMed
107Sneve, M, Figenschau, Y & Jorde, R (2008) Supplementation with cholecalciferol does not result in weight reduction in overweight and obese subjects. Eur J Endocrinol 159, 675684.Google Scholar
108Caan, B, Neuhouser, M, Aragaki, A, et al. . (2007) Calcium plus vitamin D supplementation and the risk of postmenopausal weight gain. Arch Intern Med 167, 893902.CrossRefGoogle ScholarPubMed
109Major, GC, Alarie, F, Dore, J, et al. . (2007) Supplementation with calcium+vitamin D enhances the beneficial effect of weight loss on plasma lipid and lipoprotein concentrations. Am J Clin Nutr 85, 5459.Google ScholarPubMed
110Major, GC, Alarie, FP, Dore, J, et al. . (2009) Calcium plus vitamin D supplementation and fat mass loss in female very low-calcium consumers: potential link with a calcium-specific appetite control. Br J Nutr 101, 659663.Google Scholar
111Zittermann, A, Frisch, S, Berthold, HK, et al. . (2009) Vitamin D supplementation enhances the beneficial effects of weight loss on cardiovascular disease risk markers. Am J Clin Nutr 89, 13211327.CrossRefGoogle ScholarPubMed
112Chobanian, AV, Bakris, GL, Black, HR, et al. . (2003) The Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure: the JNC 7 report. JAMA 289, 25602572.CrossRefGoogle ScholarPubMed
113Wang, Y & Wang, QJ (2004) The prevalence of prehypertension and hypertension among US adults according to the new Joint National Committee guidelines: new challenges of the old problem. Arch Intern Med 164, 21262134.Google Scholar
114Morgan, K, McGee, H, Watson, D, et al. (2008) SLÁN 2007, Survey of Lifestyle, Attitudes & Nutrition in Ireland. Main Report. Dublin: Department of Health and Children.Google Scholar
115Pilz, S, Tomaschitz, A, Ritz, E, et al. . (2009) Vitamin D status and arterial hypertension: a systematic review. Nat Rev Cardiol 6, 621630.CrossRefGoogle ScholarPubMed
116Ballermann, BJ & Marsden, PA (1991) Endothelium-derived vasoactive mediators and renal glomerular function. Clin Invest Med 14, 508517.Google Scholar
117Resnick, LM, Muller, FB & Laragh, JH (1986) Calcium-regulating hormones in essential hypertension. Relation to plasma renin activity and sodium metabolism. Ann Intern Med 105, 649654.Google Scholar
118Li, YC, Kong, J, Wei, M, et al. . (2002) 1,25-Dihydroxyvitamin D3 is a negative endocrine regulator of the renin–angiotensin system. J Clin Invest 110, 229238.Google Scholar
119Xiang, W, Kong, J, Chen, S, et al. . (2005) Cardiac hypertrophy in vitamin D receptor knockout mice: role of the systemic and cardiac renin–angiotensin systems. Am J Physiol Endocrinol Metab 288, E125E132.CrossRefGoogle ScholarPubMed
120Kong, J, Qiao, G, Zhang, Z, et al. . (2008) Targeted vitamin D receptor expression in juxtaglomerular cells suppresses renin expression independent of parathyroid hormone and calcium. Kidney Int 74, 15771581.Google Scholar
121Zhou, C, Lu, F, Cao, K, et al. . (2008) Calcium-independent and 1,25(OH)2D3-dependent regulation of the renin–angiotensin system in 1α-hydroxylase knockout mice. Kidney Int 74, 170179.Google Scholar
122Yuan, W, Pan, W, Kong, J, et al. . (2007) 1,25-Dihydroxyvitamin D3 suppresses renin gene transcription by blocking the activity of the cyclic AMP response element in the renin gene promoter. J Biol Chem 282, 2982129830.Google Scholar
123Zemel, M (2001) Calcium modulation of hypertension and obesity: mechanisms and implications. J Am Coll Nutr 20, 428435.Google Scholar
124Perkovic, V, Hewitson, TD, Kelynack, KJ, et al. . (2003) Parathyroid hormone has a prosclerotic effect on vascular smooth muscle cells. Kidney Blood Press Res 26, 2733.Google Scholar
125Rostand, SG (1997) Ultraviolet light may contribute to geographic and racial blood pressure differences. Hypertension 30, 150156.Google Scholar
126Krause, R, Buhring, M, Hopfenmuller, W, et al. . (1998) Ultraviolet B and blood pressure. Lancet 352, 709710.Google Scholar
127Sowers, MR, Wallace, RB & Lemke, JH (1985) The association of intakes of vitamin D and calcium with blood pressure among women. Am J Clin Nutr 42, 135142.Google Scholar
128Jorde, R & Bonaa, KH (2000) Calcium from dairy products, vitamin D intake, and blood pressure: the Tromso Study. Am J Clin Nutr 71, 15301535.Google Scholar
129Forman, JP, Bischoff-Ferrari, HA, Willett, WC, et al. . (2005) Vitamin D intake and risk of incident hypertension: results from three large prospective cohort studies. Hypertension 46, 676682.Google Scholar
130Wang, L, Manson, JE, Buring, JE, et al. . (2008) Dietary intake of dairy products, calcium, and vitamin D and the risk of hypertension in middle-aged and older women. Hypertension 51, 10731079.CrossRefGoogle ScholarPubMed
131Pasco, JA, Henry, MJ, Nicholson, GC, et al. . (2009) Behavioural and physical characteristics associated with vitamin D status in women. Bone 44, 10851091.Google Scholar
132Snijder, MB, Lips, P, Seidell, JC, et al. . (2007) Vitamin D status and parathyroid hormone levels in relation to blood pressure: a population-based study in older men and women. J Intern Med 261, 558565.Google Scholar
133Jorde, R, Figenschau, Y, Emaus, N, et al. . (2010) Serum 25-hydroxyvitamin D levels are strongly related to systolic blood pressure but do not predict future hypertension. Hypertension 55, 792798.Google Scholar
134Scragg, R, Sowers, M & Bell, C (2007) Serum 25-hydroxyvitamin D, ethnicity, and blood pressure in the Third National Health and Nutrition Examination Survey. Am J Hypertens 20, 713719.CrossRefGoogle ScholarPubMed
135Judd, SE, Nanes, MS, Ziegler, TR, et al. . (2008) Optimal vitamin D status attenuates the age-associated increase in systolic blood pressure in white Americans: results from the third National Health and Nutrition Examination Survey. Am J Clin Nutr 87, 136141.Google Scholar
136Martins, D, Wolf, M, Pan, D, et al. . (2007) Prevalence of cardiovascular risk factors and the serum levels of 25-hydroxyvitamin D in the United States: data from the Third National Health and Nutrition Examination Survey. Arch Intern Med 167, 11591165.Google Scholar
137Forman, JP, Giovannucci, E, Holmes, MD, et al. . (2007) Plasma 25-hydroxyvitamin D levels and risk of incident hypertension. Hypertension 49, 10631069.Google Scholar
138Forman, JP, Curhan, GC & Taylor, EN (2008) Plasma 25-hydroxyvitamin D levels and risk of incident hypertension among young women. Hypertension 52, 828832.Google Scholar
139Scragg, R, Khaw, KT & Murphy, S (1995) Effect of winter oral vitamin D3 supplementation on cardiovascular risk factors in elderly adults. Eur J Clin Nutr 49, 640646.Google Scholar
140Orwoll, ES & Oviatt, S (1990) Relationship of mineral metabolism and long-term calcium and cholecalciferol supplementation to blood pressure in normotensive men. Am J Clin Nutr 52, 717721.Google Scholar
141Margolis, KL, Ray, RM, Van Horn, L, et al. . (2008) Effect of calcium and vitamin D supplementation on blood pressure: the Women's Health Initiative Randomized Trial. Hypertension 52, 847855.Google Scholar
142Pfeifer, M, Begerow, B, Minne, HW, et al. . (2001) Effects of a short-term vitamin D3 and calcium supplementation on blood pressure and parathyroid hormone levels in elderly women. J Clin Endocrinol Metab 86, 16331637.Google Scholar
143Cox, RA & Garcia-Palmieri, MR (1990) Cholesterol, triglycerides, and associated lipoproteins. In Clinical Methods: The History, Physical and Laboratory Examinations, 3rd ed., Chapter 31, pp. 153160 [Walker, HK, Hall, WD and Hurst, JW, editors]. Stoneham, MA: Butterworth Publishers.Google Scholar
144Tziomalos, K, Athyros, VG, Wierzbicki, AS, et al. . (2009) Lipoprotein a: where are we now? Curr Opin Cardiol 24, 351357.Google Scholar
145Ballantyne, C, Arroll, B & Shepherd, J (2005) Lipids and CVD management: towards a global consensus. Eur Heart J 26, 22242231.Google Scholar
146Kendrick, J, Targher, G, Smits, G, et al. . (2009) 25-Hydroxyvitamin D deficiency is independently associated with cardiovascular disease in the Third National Health and Nutrition Examination Survey. Atherosclerosis 205, 255260.Google Scholar
147Bostom, AG, Cupples, LA, Jenner, JL, et al. . (1996) Elevated plasma lipoprotein(a) and coronary heart disease in men aged 55 years and younger. A prospective study. JAMA 276, 544548.CrossRefGoogle ScholarPubMed
148Hopkins, PN, Hunt, SC, Schreiner, PJ, et al. . (1998) Lipoprotein(a) interactions with lipid and non-lipid risk factors in patients with early onset coronary artery disease: results from the NHLBI Family Heart Study. Atherosclerosis 141, 333345.Google Scholar
149Barger-Lux, MJ, Heaney, RP, Lanspa, SJ, et al. . (1995) An investigation of sources of variation in calcium absorption efficiency. J Clin Endocrinol Metab 80, 406411.Google Scholar
150Denke, MA, Fox, MM & Schulte, MC (1993) Short-term dietary calcium fortification increases fecal saturated fat content and reduces serum lipids in men. J Nutr 123, 10471053.Google Scholar
151Reid, IR (2004) Effects of calcium supplementation on circulating lipids: potential pharmacoeconomic implications. Drugs Aging 21, 717.Google Scholar
152Vaskonen, T, Mervaala, E, Sumuvuori, V, et al. . (2002) Effects of calcium and plant sterols on serum lipids in obese Zucker rats on a low-fat diet. Br J Nutr 87, 239245.CrossRefGoogle ScholarPubMed
153Cho, HJ, Kang, HC, Choi, SA, et al. . (2005) The possible role of Ca2+ on the activation of microsomal triglyceride transfer protein in rat hepatocytes. Biol Pharm Bull 28, 14181423.Google Scholar
154Lacour, B, Basile, C, Drueke, T, et al. . (1982) Parathyroid function and lipid metabolism in the rat. Miner Electrolyte Metab 7, 157165.Google ScholarPubMed
155Kelly, GS (2005) Seasonal variations of selected cardiovascular risk factors. Altern Med Rev 10, 307320.Google ScholarPubMed
156Cigolini, M, Iagulli, MP, Miconi, V, et al. . (2006) Serum 25-hydroxyvitamin D3 concentrations and prevalence of cardiovascular disease among type 2 diabetic patients. Diabetes Care 29, 722724.CrossRefGoogle ScholarPubMed
157Auwerx, J, Bouillon, R & Kesteloot, H (1992) Relation between 25-hydroxyvitamin D3, apolipoprotein A-I, and high density lipoprotein cholesterol. Arterioscler Thromb 12, 671674.Google Scholar
158Carbone, LD, Rosenberg, EW, Tolley, EA, et al. . (2008) 25-Hydroxyvitamin D, cholesterol, and ultraviolet irradiation. Metabolism 57, 741748.Google Scholar
159John, WG, Noonan, K, Mannan, N, et al. . (2005) Hypovitaminosis D is associated with reductions in serum apolipoprotein A-I but not with fasting lipids in British Bangladeshis. Am J Clin Nutr 82, 517522.Google Scholar
160Wang, TJ, Pencina, MJ, Booth, SL, et al. . (2008) Vitamin D deficiency and risk of cardiovascular disease. Circulation 117, 503511.Google Scholar
161Andersen, R, Brot, C, Mejborn, H, et al. . (2009) Vitamin D supplementation does not affect serum lipids and lipoproteins in Pakistani immigrants. Eur J Clin Nutr 63, 11501153.Google Scholar
162Gannage-Yared, MH, Azoury, M, Mansour, I, et al. . (2003) Effects of a short-term calcium and vitamin D treatment on serum cytokines, bone markers, insulin and lipid concentrations in healthy post-menopausal women. J Endocrinol Invest 26, 748753.Google Scholar
163Rajpathak, SN, Xue, X, Wassertheil-Smoller, S, et al. . (2010) Effect of 5 y of calcium plus vitamin D supplementation on change in circulating lipids: results from the Women's Health Initiative. Am J Clin Nutr 91, 894899.Google Scholar
164Daly, RM & Nowson, CA (2009) Long-term effect of calcium–vitamin D3 fortified milk on blood pressure and serum lipid concentrations in healthy older men. Eur J Clin Nutr 63, 9931000.Google Scholar
165Osmancevic, A, Nilsen, LT, Landin-Wilhelmsen, K, et al. . (2009) Effect of climate therapy at Gran Canaria on vitamin D production, blood glucose and lipids in patients with psoriasis. J Eur Acad Dermatol Venereol 23, 11331140.Google Scholar
166Tuppurainen, M, Heikkinen, AM, Penttilä, I, et al. . (1995) Does vitamin D3 have negative effects on serum levels of lipids? A follow-up study with a sequential combination of estradiol valerate and cyproterone acetate and/or vitamin D3. Maturitas 22, 5561.Google Scholar
167Heikkinen, AM, Tuppurainen, MT, Niskanen, L, et al. . (1997) Long-term vitamin D3 supplementation may have adverse effects on serum lipids during postmenopausal hormone replacement therapy. Eur J Endocrinol 137, 495502.Google Scholar
168Wang, JH, Keisala, T, Solakivi, T, et al. . (2009) Serum cholesterol and expression of ApoAI, LXRβ and SREBP2 in vitamin D receptor knock-out mice. J Steroid Biochem Mol Biol 113, 222226.Google Scholar
169Wehmeier, K, Beers, A, Haas, MJ, et al. . (2005) Inhibition of apolipoprotein AI gene expression by 1, 25-dihydroxyvitamin D3. Biochim Biophys Acta 1737, 1626.Google Scholar
170Kowalska, I, Straczkowski, M, Nikolajuk, A, et al. . (2008) Insulin resistance, serum adiponectin, and proinflammatory markers in young subjects with the metabolic syndrome. Metabolism 57, 15391544.CrossRefGoogle ScholarPubMed
171Libby, P (2002) Inflammation in atherosclerosis. Nature 420, 868874.Google Scholar
172Libby, P, Ridker, PM & Maseri, A (2002) Inflammation and atherosclerosis. Circulation 105, 11351143.Google Scholar
173Stentz, FB, Umpierrez, GE, Cuervo, R, et al. . (2004) Proinflammatory cytokines, markers of cardiovascular risks, oxidative stress, and lipid peroxidation in patients with hyperglycemic crises. Diabetes 53, 20792086.Google Scholar
174Moore, K, de Waal Malefyt, R, Coffman, R, et al. . (2001) Interleukin-10 and the interleukin-10 receptor. Annu Rev Immunol 19, 683765.Google Scholar
175Festa, A, D'Agostino, R Jr, Howard, G, et al. . (2000) Chronic subclinical inflammation as part of the insulin resistance syndrome: the Insulin Resistance Atherosclerosis Study (IRAS). Circulation 102, 42.CrossRefGoogle ScholarPubMed
176Ridker, PM, Buring, JE, Cook, NR, et al. . (2003) C-reactive protein, the metabolic syndrome, and risk of incident cardiovascular events: an 8-year follow-up of 14 719 initially healthy American women. Circulation 107, 391397.Google Scholar
177Adams, JS & Hewison, M (2008) Unexpected actions of vitamin D: new perspectives on the regulation of innate and adaptive immunity. Nat Clin Pract Endocrinol Metab 4, 8090.Google Scholar
178Cantorna, MT, Zhu, Y, Froicu, M, et al. . (2004) Vitamin D status, 1,25-dihydroxyvitamin D3, and the immune system. Am J Clin Nutr 80, 1717S1720S.Google Scholar
179Herder, C, Baumert, J, Thorand, B, et al. . (2006) Chemokines as risk factors for type 2 diabetes: results from the MONICA/KORA Augsburg study, 1984–2002. Diabetologia 49, 921929.Google Scholar
180Deluca, HF & Cantorna, MT (2001) Vitamin D: its role and uses in immunology. FASEB J 15, 25792585.Google Scholar
181Mathieu, C & Adorini, L (2002) The coming of age of 1,25-dihydroxyvitamin D3 analogs as immunomodulatory agents. Trends Mol Med 8, 174179.CrossRefGoogle ScholarPubMed
182Overbergh, L, Decallonne, B, Valckx, D, et al. . (2000) Identification and immune regulation of 25-hydroxyvitamin D-1-α-hydroxylase in murine macrophages. Clin Exp Immunol 120, 139.Google Scholar
183van Etten, E & Mathieu, C (2005) Immunoregulation by 1, 25-dihydroxyvitamin D3: basic concepts. J Steroid Biochem Mol Biol 97, 93101.Google Scholar
184van Etten, E, Stoffels, K, Gysemans, C, et al. . (2008) Regulation of vitamin D homeostasis: implications for the immune system. Nutr Rev 66, Suppl. 2, 125134.Google Scholar
185Zhu, Y, Mahon, BD, Froicu, M, et al. . (2005) Calcium and 1 α,25-dihydroxyvitamin D3 target the TNF-α pathway to suppress experimental inflammatory bowel disease. Eur J Immunol 35, 217224.Google Scholar
186Talmor-Barkan, Y, Rashid, G, Weintal, I, et al. . (2009) Low extracellular Ca2+: a mediator of endothelial inflammation. Nephrol Dial Transplant 24, 33063312.Google Scholar
187Greenfield, EM, Gornik, SA, Horowitz, MC, et al. . (1993) Regulation of cytokine expression in osteoblasts by parathyroid hormone: rapid stimulation of interleukin-6 and leukemia inhibitory factor mRNA. J Bone Miner Res 8, 11631171.Google Scholar
188Shea, M, Booth, S, Massaro, J, et al. . (2007) Vitamin K and vitamin D status: associations with inflammatory markers in the Framingham Offspring Study. Am J Epidemiol 167, 313320.Google Scholar
189Peterson, C & Heffernan, M (2008) Serum tumor necrosis factor-α concentrations are negatively correlated with serum 25 (OH) D concentrations in healthy women. J Inflamm (Lond) 5, 10.Google Scholar
190Van den Berghe, G, Van Roosbroeck, D, Vanhove, P, et al. . (2003) Bone turnover in prolonged critical illness: effect of vitamin D. J Clin Endocrinol Metab 88, 46234632.CrossRefGoogle ScholarPubMed
191Schleithoff, SS, Zittermann, A, Tenderich, G, et al. . (2006) Vitamin D supplementation improves cytokine profiles in patients with congestive heart failure: a double-blind, randomized, placebo-controlled trial. Am J Clin Nutr 83, 754759.CrossRefGoogle ScholarPubMed
192Mahon, B, Gordon, S, Cruz, J, et al. . (2003) Cytokine profile in patients with multiple sclerosis following vitamin D supplementation. J Neuroimmunol 134, 128132.Google Scholar
193Jorde, R, Sneve, M, Torjesen, PA, et al. . (2010) No effect of supplementation with cholecalciferol on cytokines and markers of inflammation in overweight and obese subjects. Cytokine 50, 175180.CrossRefGoogle Scholar
194Weinreich, T, Wuthrich, RP, Booy, C, et al. . (2001) Suppression of ICAM-1 expression in renal proximal tubular cells by 1,25-dihydroxyvitamin D3. Kidney Blood Press Res 24, 9298.Google Scholar
195Martinesi, M, Bruni, S, Stio, M, et al. . (2006) 1,25-Dihydroxyvitamin D3 inhibits tumor necrosis factor-α-induced adhesion molecule expression in endothelial cells. Cell Biol Int 30, 365375.Google Scholar
196Si-Feng, C (1995) 1α,25-Dihydroxyvitamin D3 decreased ICAM-1 and ELAM-1 expressions on pulmonary microvascular endothelial cells and neutrophil motivation. J Steroid Biochem Mol Biol 52, 6770.Google Scholar
197Kreft, B, Brzoska, S, Doehn, C, et al. . (1996) 1,25-Dihydroxycholecalciferol enhances the expression of MHC class II antigens and intercellular adhesion molecule-1 by human renal tubular epithelial cells. J Urol 155, 14481453.Google Scholar
198Kohler, HP & Grant, PJ (2000) Plasminogen-activator inhibitor type 1 and coronary artery disease. N Engl J Med 342, 17921801.CrossRefGoogle ScholarPubMed
199Thogersen, A, Jansson, J, Boman, K, et al. . (1998) High plasminogen activator inhibitor and tissue plasminogen activator levels in plasma precede a first acute myocardial infarction in both men and women: evidence for the fibrinolytic system as an independent primary risk factor. Circulation 98, 2241.Google Scholar
200Hamsten, A, Wiman, B, de Faire, U, et al. . (1985) Increased plasma levels of a rapid inhibitor of tissue plasminogen activator in young survivors of myocardial infarction. N Engl J Med 313, 15571563.Google Scholar
201Hamsten, A, de Faire, U, Walldius, G, et al. . (1987) Plasminogen activator inhibitor in plasma: risk factor for recurrent myocardial infarction. Lancet ii, 39.Google Scholar
202Wu-Wong, JR, Nakane, M & Ma, J (2007) Vitamin D analogs modulate the expression of plasminogen activator inhibitor-1, thrombospondin-1 and thrombomodulin in human aortic smooth muscle cells. J Vasc Res 44, 1118.Google Scholar
203Wu-Wong, JR, Nakane, M, Ma, J, et al. . (2006) Effects of vitamin D analogs on gene expression profiling in human coronary artery smooth muscle cells. Atherosclerosis 186, 2028.Google Scholar
204Vaughan, DE, Lazos, SA & Tong, K (1995) Angiotensin II regulates the expression of plasminogen activator inhibitor-1 in cultured endothelial cells. A potential link between the renin-angiotensin system and thrombosis. J Clin Invest 95, 9951001.CrossRefGoogle ScholarPubMed
205Brown, NJ, Agirbasli, M & Vaughan, DE (1999) Comparative effect of angiotensin-converting enzyme inhibition and angiotensin II type 1 receptor antagonism on plasma fibrinolytic balance in humans. Hypertension 34, 285290.Google Scholar
206Jorde, R, Sneve, M, Torjesen, P, et al. . (2010) Parameters of the thrombogram are associated with serum 25-hydroxyvitamin D levels at baseline, but not affected during supplementation with vitamin D. Thromb Res 125, e210e213.Google Scholar
207Rahman, A, Hershey, S, Ahmed, S, et al. . (2007) Heart extracellular matrix gene expression profile in the vitamin D receptor knockout mice. J Steroid Biochem Mol Biol 103, 416419.Google Scholar
208Dollery, C & Libby, P (2006) Atherosclerosis and proteinase activation. Cardiovasc Res 69, 625.Google Scholar
209Dean, D, Schwartz, Z, Schmitz, J, et al. . (1996) Vitamin D regulation of metalloproteinase activity in matrix vesicles. Connect Tissue Res 35, 331336.Google Scholar
210Tai, K, Need, AG, Horowitz, M, et al. . (2008) Glucose tolerance and vitamin D: effects of treating vitamin D deficiency. Nutrition 24, 950956.Google Scholar
211Raghuramulu, N, Raghunath, M, Chandra, S, et al. . (1992) Vitamin D improves oral glucose tolerance and insulin secretion in human diabetes. J Clin Biochem Nutr 13, 4551.Google Scholar
Figure 0

Table 1 The effect of cholecalciferol supplementation (as a single treatment) on cardiometabolic biomarkers