Hostname: page-component-7c8c6479df-8mjnm Total loading time: 0 Render date: 2024-03-28T17:09:15.876Z Has data issue: false hasContentIssue false

Old wine in new bottles: vitamin D in the treatment and prevention of tuberculosis

Published online by Cambridge University Press:  29 November 2011

Adrian R. Martineau*
Affiliation:
Queen Mary University of London, Barts and The London School of Medicine and Dentistry, London E1 2AB, UK
*
Corresponding author: Dr Adrian Martineau, fax +44 207 882 2552, email a.martineau@qmul.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

Tuberculosis (TB) is a major cause of mortality, responsible for 1·68 million deaths worldwide in 2009. The global prevalence of latent Mycobacterium tuberculosis infection is estimated to be 32%, and this carries a 5–20% lifetime risk of reactivation disease. The emergence of drug-resistant organisms necessitates the development of new agents to enhance the response to antimicrobial therapy for active TB. Vitamin D was used to treat TB in the pre-antibiotic era, and its active metabolite, 1,25-dihydoxyvitamin D, has long been known to enhance the immune response to mycobacteria in vitro. Vitamin D deficiency is common in patients with active TB, and several clinical trials have evaluated the role of adjunctive vitamin D supplementation in its treatment. Results of these studies are conflicting, reflecting variation between studies in baseline vitamin D status of participants, dosing regimens and outcome measures. Vitamin D deficiency is also recognised to be highly prevalent among people with latent M. tuberculosis infection in both high- and low-burden settings, and there is a wealth of observational epidemiological evidence linking vitamin D deficiency with increased risk of reactivation disease. Randomised controlled trials of vitamin D supplementation for the prevention of active TB have yet to be performed, however. The conduct of such trials is a research priority, given the safety and low cost of vitamin D supplementation, and the potentially huge public health consequences of positive results.

Type
70th Anniversary Conference on ‘Vitamins in early development and healthy aging: impact on infectious and chronic disease’
Copyright
Copyright © The Author 2011

Abbreviations:
1,25(OH)2D

1,25-dihydoxyvitamin D

25(OH)D

25-hydroxyvitamin D

DBP

vitamin D binding protein

Gc2

group-specific component 2

MTB

Mycobacterium tuberculosis

TB

tuberculosis

VDR

vitamin D receptor

Tuberculosis (TB) is a major public health problem. The global prevalence of latent Mycobacterium tuberculosis (MTB) infection has been estimated at 32%( Reference Dye, Scheele and Dolin 1 ), and this carries a 5–20% lifetime risk of reactivation disease in people who are not infected with HIV( Reference Horsburgh 2 ); reactivation rates higher than 10% per annum have been reported in HIV-infected people( Reference Wood, Maartens and Lombard 3 ). The WHO estimates that in 2009 there were 9·4 million incident cases of active TB, 14 million prevalent cases of TB, 1·3 million deaths from TB in HIV-uninfected people and 0·38 million deaths from TB in HIV-infected people( 4 ). The development of new agents to prevent acquisition or reactivation of latent MTB infection and to allow shortening of antimicrobial therapy regimens for active TB without loss of efficacy is a research priority. This paper reviews the growing body of evidence from studies conducted both in vitro and in vivo suggesting that vitamin D might have a role in the prevention and treatment of TB.

Immunomodulatory actions of vitamin D in mycobacterial infection

With the exception of a single report( Reference Raab 5 ), vitamin D and its metabolites have not been shown to possess antimycobacterial activity in the absence of cells. However, the active metabolite of vitamin D, 1,25-dihydoxyvitamin D (1,25(OH)2D), has long been recognised to induce antimycobacterial activity in vitro in mononuclear phagocytes, the cells that control growth of MTB( Reference Rook, Steele and Fraher 6 ). Ligation of macrophage Toll-like receptor 2/1 heterodimers by mycobacterial antigens induces expression of the vitamin D receptor (VDR) and the 1-α hydroxylase enzyme CYP27B1( Reference Liu, Stenger and Li 7 , Reference Krutzik, Hewison and Liu 8 ), which synthesises 1,25(OH)2D from the principal circulating vitamin D metabolite 25-hydroxyvitamin D (25(OH)D). Because extra-renal 1-α hydroxylase follows first-order kinetics, the rate at which it synthesises 1,25(OH)2D depends on the availability of 25(OH)D substrate( Reference Vieth, McCarten and Norwich 9 ). Orally ingested vitamin D is freely converted to 25(OH)D( Reference Vieth, Glorieux, Pike and Feldman 10 ), and this provides the rationale for administering ‘parent’ vitamin D to induce antimycobacterial responses at the site of disease.

1,25(OH)2D modulates immune responses by ligating membrane VDR to induce rapid effects (within minutes) or nuclear VDR to induce genomic effects (within hours)( Reference Norman, Mizwicki and Norman 11 ). Experiments using selective agonists and antagonists of these two receptors indicate that ligation of nuclear VDR is both necessary and sufficient for induction of antimycobacterial responses by 1,25(OH)2D in vitro ( Reference Martineau, Wilkinson and Newton 12 ). 1,25(OH)2D modulates the host response to mycobacterial infection by pleiotropic mechanisms including the induction of reactive nitrogen and oxygen intermediates( Reference Rockett, Brookes and Udalova 13 , Reference Sly, Lopez and Nauseef 14 ), down-regulation of the gene encoding tryptophan-aspartate containing coat protein( Reference Anand and Kaul 15 ), promotion of phagolysosome fusion( Reference Hmama, Sendide and Talal 16 ), suppression of matrix metalloproteinase enzymes implicated in the pathogenesis of pulmonary cavitation( Reference Coussens, Timms and Boucher 17 ) and induction of antimicrobial peptides including cathelicidin LL-37( Reference Liu, Stenger and Li 7 , Reference Martineau, Wilkinson and Newton 12 ) and human β-defensin 2( Reference Liu, Schenk and Walker 18 ). Cathelicidin LL-37 possesses antimycobacterial activity( Reference Liu, Stenger and Li 7 , Reference Martineau, Newton and Wilkinson 19 ) and also induces autophagy( Reference Yuk, Shin and Lee 20 , Reference Shin, Yuk and Lee 21 ); 1,25(OH)2D3-induced antimycobacterial activity has been reported to be dependent on expression of the gene encoding cathelicidin LL-37( Reference Liu, Stenger and Tang 22 ).

Vitamin D and tuberculosis: historical aspects

The clinical features of vitamin D deficiency were first described in 1651, when Glisson, Bate and Regemorter published ‘A treatise of the rickets: being a disease common to children’( Reference Glisson, Bate and Regemorter 23 ). In addition to noting the classical musculoskeletal features of rickets, the authors made the following observation from an autopsy of an infant with the condition: ‘One amongst us doth attest, that he saw glandulous knobs and bunches so numerous that they seemed to equalise, if not exceed, the magnitude of the lungs themselves; they were situated between the lungs and the mediastinum … and were extended from the Canel bone to the Diaphragma’. TB is a well-recognised cause of mediastinal lymphadenopathy in children( Reference De Ugarte, Shapiro and Williams 24 ) and it is interesting to speculate whether this represents the earliest case report of TB associated with vitamin D deficiency.

Some 200 years later, Chapman reported results of administering cod liver oil to patients with ‘consumption’, and made the following observation: ‘the beneficial effects of the oil were manifested most speedily and most decisively, in the improvement of the appetite, aspect of the countenance, strength and spirits’. He concluded that cod liver oil was ‘probably the only remedial agent by which the vital powers may be enabled to struggle successfully against that malady’( Reference Chapman 25 ). This report represents the first circumstantial evidence that administration of a preparation containing vitamin D improved clinical outcome in patients with TB, although it should be noted that no control group was studied, and that any beneficial effects of cod liver oil may have been attributable to its content of vitamin A rather than vitamin D( Reference Karyadi, West and Schultink 26 ). The first TB sanatorium was opened in Gorbersdorf, Germany (today Sokolowsko, Poland) in 1859, and heliotherapy (exposure of TB patients to sunlight, which induces cutaneous vitamin D synthesis) subsequently became common practice, and was credited with improvements in clinical outcome in many cases( Reference Mayer 27 ). In 1903, Niels Finsen was awarded the Nobel Prize in Physiology or Medicine for his discovery that shortwave UV light was effective in the treatment of cutaneous TB( Reference Roelandts 28 ). Vitamin D2 was purified and crystallised in 1931( Reference Askew, Bruce and Callow 29 ) and Charpy subsequently pioneered the use of pharmacologic doses (≥1·25 mg daily) of vitamin D2 to treat cutaneous TB( Reference Charpy 30 ). Vitamin D2 was also used to treat pulmonary TB, both as a single agent and, following the introduction of effective anti-TB chemotherapy, as an adjunct to antibiotic treatment( Reference Martineau, Honecker and Wilkinson 31 ).

Association between vitamin D deficiency and susceptibility to tuberculosis

In 1985, Davies observed that people migrating to the United Kingdom from countries with a high incidence of latent MTB infection experienced rates of active TB that exceeded rates in their countries of origin, and that this increased risk coincided with the development of vitamin D deficiency, probably arising as a result of decreased sun exposure( Reference Davies 32 ). He suggested that vitamin D deficiency may predispose to reactivation of latent MTB infection in this setting, a hypothesis supported by his observation that vitamin D deficiency associated with susceptibility to active TB( Reference Davies, Brown and Woodhead 33 ). Since then, eleven case–control studies investigating the association between vitamin D status and susceptibility to active TB have been published. Of these, seven have reported a statistically significant association between vitamin D deficiency and susceptibility to active TB( Reference Davies, Church and Brown 34 Reference Ho-Pham, Nguyen and Nguyen 40 ), three have reported a non-statistically significant trend towards such an association( Reference Grange, Davies and Brown 41 Reference Martineau, Leandro and Anderson 43 ) and one( Reference Nielsen, Skifte and Andersson 44 ) has reported that active TB was associated with both ‘high’ and ‘low’ serum 25(OH)D concentrations (>140 and <75 nmol/l, respectively). Potential explanations for an association between vitamin D deficiency and active TB include both causality (i.e. vitamin D deficiency impairs host immune response to MTB and causes susceptibility) and reverse causality (i.e. active TB causes vitamin D deficiency, due to anorexia, decreased exposure to sunlight in debilitated patients, or MTB-induced dysregulation of vitamin D metabolism( Reference Sita-Lumsden, Lapthorn and Swaminathan 38 )).

Association between susceptibility to tuberculosis and polymorphisms in the vitamin D receptor and vitamin D binding protein

Human VDR is encoded by the VDR gene located on chromosome 12q. This gene is polymorphic, and numerous SNP have been described. The hypothesis that VDR variants might associate with susceptibility to active TB was first investigated by Bellamy et al., who reported an association between carriage of the T allele of the TaqI VDR polymorphism and susceptibility to active TB in a case–control study conducted in Gambian adults( Reference Bellamy, Ruwende and Corrah 45 ). Wilkinson et al. subsequently reported that associations between susceptibility to TB and carriage of the T allele of the TaqI VDR polymorphism and the ff genotype of the FokI VDR polymorphism in Gujarati Asians living in London were restricted to vitamin D-deficient individuals( Reference Wilkinson, Llewelyn and Toossi 36 ); this study is the first to report that gene–environment interactions may operate to influence susceptibility to active TB. Numerous case–control studies investigating the association between VDR variants and susceptibility to active TB have since been published; a recent meta-analysis of twenty-three such studies reported that in Asian populations, the FokI ff genotype associated with susceptibility to active TB (pooled OR 2·0, 95% CI 1·3, 3·2), and the BsmI bb genotype (defined by the presence of two restriction sites for the Bsm1 endonuclease) was associated with protection against active TB (pooled OR 0·5, 95% CI 0·4, 0·8); no associations were seen in African or Latin American populations, however( Reference Gao, Tao and Zhang 46 ).

Further case–control studies have investigated associations between polymorphisms in the vitamin D binding protein (DBP) and susceptibility to active TB. DBP is a highly expressed multifunctional 58 kDa serum glycoprotein encoded on chromosome 4. Two common polymorphisms at codons 416 and 420 of exon 11 of the DBP gene give rise to the three major electrophoretic variants of DBP, termed group-specific component 1 fast, group-specific component 1 slow and group-specific component 2 (Gc2). These variants differ in their functional characteristics: the group-specific component 1 fast and group-specific component 1 slow variants have been reported to have greater affinity for 25(OH)D than the Gc2 variant( Reference Arnaud and Constans 47 ), potentially leading to more efficient delivery of 25(OH)D to the target tissues, while the Gc2 variant is associated with decreased circulating concentrations of 25(OH)D, 1,25(OH)2D and DBP( Reference Lauridsen, Vestergaard and Hermann 48 , Reference Abbas, Linseisen and Slanger 49 ). Case–control studies conducted in India, Russia and Kuwait have not reported any association between DBP genotype and susceptibility to TB( Reference Papiha, Agarwal and White 50 Reference Bahr, Eales and Nye 52 ), but a more recent study reported an association between the Gc2 allele of DBP and susceptibility to active TB among Gujarati Asians living in London. This association was preserved if serum 25(OH)D concentration was <20 nmol/l, but not if serum 25(OH)D was ≥20 nmol/l, suggesting that profound vitamin D deficiency and Gc2 genotype may interact to increase susceptibility to TB( Reference Martineau, Leandro and Anderson 43 ).

Prospective observational studies

In contrast to the numbers of published cross-sectional studies, relatively few cohort studies investigating associations between vitamin D status or VDR genotype and TB have been conducted. Two studies have examined the influence of VDR genotype on response to antimicrobial therapy: Roth et al. reported that the FF genotype of the FokI VDR polymorphism and the Tt genotype of the TaqI VDR polymorphism associated with faster sputum culture conversion in a cohort of pulmonary TB patients in Peru( Reference Roth, Soto and Arenas 53 ), while Babb et al. reported no difference in time to sputum culture conversion according to TaqI or FokI VDR genotype among South African TB patients( Reference Babb, van der Merwe and Beyers 54 ). Recently, a cohort study conducted in Pakistan( Reference Talat, Perry and Parsonnet 55 ) reported that profound vitamin D deficiency among healthy household TB contacts at baseline associated with increased risk of development of active TB over the subsequent 4 years: seven out of thirty contacts with baseline plasma 25(OH)D <17·5 nmol/l developed active TB during follow-up, compared with one of thirty-two with plasma 25(OH)D 17·5–33·5 nmol/l and none of thirty with plasma 25(OH)D >33·5 nmol/l. This association retained significance after adjustment for age and sex, although other potential confounders were not taken into account in the analysis. The observation that increased risk of TB reactivation was almost exclusively confined to individuals with profound vitamin D deficiency is interesting, particularly when taken together with reports from case–control studies that profound vitamin D deficiency is most strongly associated with susceptibility to TB( Reference Wilkinson, Llewelyn and Toossi 36 ): the implication is that, if vitamin D deficiency does indeed predispose to active TB, then relatively modest elevations of serum 25(OH)D might be effective for the prevention of active disease.

Intervention studies

Despite the compelling results from the laboratory and observational studies reviewed above, no randomised controlled trials of vitamin D supplementation for the prevention of active TB have been published to date. The absence of such studies reflects the very considerable methodological and logistic challenges of conducting them. Because the annual risk of reactivation of latent TB is low in immunocompetent individuals (<1% even in individuals with a strongly positive and newly converted tuberculin skin test( Reference Horsburgh 2 )), very large sample sizes and prolonged follow-up will be needed to detect all but the largest effects of vitamin D supplementation on TB incidence in such populations. One study has attempted to circumvent this problem by investigating the effect of vitamin D supplementation on a surrogate outcome measure of antimycobacterial response: the BCG-lux assay, which measures the ability of whole blood to restrict bioluminescence of a reporter mycobacterium( Reference Kampmann, Gaora and Snewin 56 ). This investigation found that a single dose of 2·5 mg vitamin D enhanced the ability of TB contacts’ whole blood to restrict mycobacterial bioluminescence at 24 h post-inoculation( Reference Martineau, Wilkinson and Wilkinson 57 ), providing further evidence that trials of vitamin D supplementation for the prevention of TB are justified.

In contrast to prevention studies, randomised controlled trials to determine whether adjunctive vitamin D enhances response to antimicrobial therapy can be powered with more modest numbers of participants, because the majority of TB patients respond rapidly to antimicrobial therapy. Seven such studies have been published to date (summarised in Table 1). Three of these trials had biochemical primary outcomes: two reported no hypercalcaemia in TB patients receiving either 125 μg vitamin D daily( Reference Gwinup, Randazzo and Elias 58 ) or a single oral dose of 2·5 mg vitamin D( Reference Martineau, Nanzer and Satkunam 59 ), and one reported hypercalcaemia occurring in nineteen of thirty TB patients receiving daily doses of 10–95 μg vitamin D( Reference Narang, Gupta and Jain 60 ). However, this third study, by Narang et al., also reported that a daily dose of 60 μg vitamin D elevated mean serum Ca in healthy controls; a finding that contrasts with other studies which demonstrate that identical( Reference Tjellesen, Hummer and Christiansen 61 ) or considerably higher( Reference Stern, Taylor and Bell 62 ) doses of vitamin D do not induce hypercalcaemia in healthy people. It is possible, therefore, that the actual doses of vitamin D administered in Narang's study were considerably higher than reported. The remaining four clinical trials listed in the table had clinical primary outcomes. Morcos et al. investigated the effects of 25 μg vitamin D daily on twenty-four children in Egypt receiving antimicrobial therapy for TB, and showed no effect on body weight or resolution of symptoms( Reference Morcos, Gabr and Samuel 63 ). Nursyam et al. subsequently conducted a trial of a daily dose of 250 μg vitamin D in sixty-seven pulmonary TB patients in Indonesia( Reference Nursyam, Amin and Rumende 64 ). In this study, adjunctive vitamin D enhanced sputum smear conversion at 6 weeks after initiation of antimicrobial therapy (thirty-four out of thirty-four v. twenty-five out of thirty-three smear-converted in intervention v. control arm at 6 weeks, P=0·002); no effect of the intervention was seen at 8 weeks. The vitamin D status of participants was not assessed at either baseline or follow-up in this study, and details of safety monitoring, including monitoring of serum Ca concentrations, were not reported. In the largest treatment trial published to date, Wejse et al. randomised 365 adult TB patients in Guinea–Bissau to receive three doses of 2·5 mg vitamin D3 or placebo at initiation of antimicrobial therapy, and again at 5 and 8 months( Reference Wejse, Gomes and Rabna 65 ). The intervention had no effect on the primary outcome measure (a specially designed TB score) or on serum 25(OH)D concentration. Mean serum 25(OH)D concentrations at baseline were 78 nmol/l v. 79 nmol/l in intervention v. control groups. Most recently, another trial investigated the effect of a 2-weekly dose of 2·5 mg vitamin D on time to sputum culture conversion in 146 patients with smear-positive pulmonary TB in the UK( Reference Martineau, Timms and Bothamley 66 ). A 79 nmol/l increase in serum 25(OH)D was seen among participants in the intervention arm of the study, which was associated with a non-statistically significant trend towards faster sputum culture conversion (P=0·14). A pre-planned subgroup analysis revealed that adjunctive vitamin D significantly hastened sputum culture conversion by more than 17 d in participants with the tt genotype of the TaqI VDR polymorphism (hazard ratio 8·09, 95% CI 1·36, 48·01; P=0·02).

Table 1. Summary of randomised controlled trials investigating effects of adjunctive vitamin D in patients with tuberculosis (TB)

* Type of vitamin D (D2 v. D3) not reported.

Conclusions

Much remains to be done to evaluate whether vitamin D might have a role in the prevention or treatment of TB. A key research priority is to establish randomised controlled trials of vitamin D supplementation for the prevention of TB in individuals with latent MTB infection. Although some question the need for such studies to be conducted on the grounds that data from observational studies are suggestive, and that the methodological challenges of conducting such trials are too great( Reference Vieth 67 ), I remain convinced that these trials are necessary, fundable and feasible. Equivalent trials have been conducted to establish the role of chemoprophylaxis for TB prevention( Reference Ferebee 68 ), and investigation of the role of vitamin D supplementation in this regard should be no less of a research priority, given the safety and low cost of this intervention. Investigations of the potential role of vitamin D as an adjunct to antimicrobial therapy are more advanced, but results from clinical trials published to date have shown little if any benefit in drug-sensitive disease. This is not the end of the road for this line of enquiry, however. First, five other similar trials are ongoing( Reference Vieth 67 ); a meta-analysis of the results of these studies may reveal a benefit that existing studies have not been powered to demonstrate. Second, the doses of vitamin D administered in trials conducted to date are considerably lower than those reported to be effective historically( Reference Martineau, Honecker and Wilkinson 31 ); the effects of pharmacological dosing regimens are worthy of investigation. Finally, on-going investigations from a recently completed trial( Reference Martineau, Timms and Bothamley 66 ) reveal that administration of adjunctive vitamin D is associated with favourable immunomodulatory activity; this observation raises the possibility that individuals with multi-drug resistant TB, in whom antimicrobial therapy is less effective, might derive a clinical benefit from enhancement of their antimycobacterial immune response using adjunctive vitamin D therapy.

Acknowledgements

The author declares no conflict of interest. This work received no specific grant from any funding agency in the public, commercial or not-for-profit sectors.

References

1. Dye, C, Scheele, S, Dolin, P et al. (1999) Consensus statement. Global burden of tuberculosis: Estimated incidence, prevalence, and mortality by country. WHO Global Surveillance and Monitoring Project. JAMA 282, 677686.CrossRefGoogle ScholarPubMed
2. Horsburgh, CR Jr (2004) Priorities for the treatment of latent tuberculosis infection in the United States. N Engl J Med 350, 20602067.CrossRefGoogle ScholarPubMed
3. Wood, R, Maartens, G & Lombard, CJ (2000) Risk factors for developing tuberculosis in HIV-1-infected adults from communities with a low or very high incidence of tuberculosis. J Acquir Immune Defic Syndr 23, 7580.CrossRefGoogle ScholarPubMed
4. WHO (2010) Global Tuberculosis Control: WHO Report 2010. Geneva: WHO Press.Google Scholar
5. Raab, W (1946) Vitamin D – its bactericidal action. Chest 12, 409415.Google ScholarPubMed
6. Rook, GA, Steele, J, Fraher, L et al. (1986) Vitamin D3, gamma interferon, and control of proliferation of Mycobacterium tuberculosis by human monocytes. Immunology 57, 159163.Google ScholarPubMed
7. Liu, PT, Stenger, S, Li, H et al. (2006) Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response. Science 311, 17701773.CrossRefGoogle ScholarPubMed
8. Krutzik, SR, Hewison, M, Liu, PT et al. (2008) IL-15 links TLR2/1-induced macrophage differentiation to the vitamin D-dependent antimicrobial pathway. J Immunol 181, 71157120.CrossRefGoogle Scholar
9. Vieth, R, McCarten, K & Norwich, KH (1990) Role of 25-hydroxyvitamin D3 dose in determining rat 1,25-dihydroxyvitamin D3 production. Am J Physiol 258, E780E789.Google Scholar
10. Vieth, R (2005) The pharmacology of vitamin D, including fortification strategies. In Vitamin D pp. 995–1015 [Glorieux, FH, Pike, JW and Feldman, D]. London: Academic Press.Google ScholarPubMed
11. Norman, AW, Mizwicki, MT & Norman, DP (2004) Steroid-hormone rapid actions, membrane receptors and a conformational ensemble model. Nat Rev Drug Discov 3, 2741.CrossRefGoogle Scholar
12. Martineau, AR, Wilkinson, KA, Newton, SM et al. (2007) IFN-γ- and TNF-independent vitamin D-inducible human suppression of mycobacteria: The role of cathelicidin LL-37. J Immunol 178, 71907198.CrossRefGoogle ScholarPubMed
13. Rockett, KA, Brookes, R, Udalova, I et al. (1998) 1,25-dihydroxyvitamin D3 induces nitric oxide synthase and suppresses growth of Mycobacterium tuberculosis in a human macrophage-like cell line. Infect Immun 66, 53145321.CrossRefGoogle Scholar
14. Sly, LM, Lopez, M, Nauseef, WM et al. (2001) 1alpha,25-dihydroxyvitamin D3-induced monocyte antimycobacterial activity is regulated by phosphatidylinositol 3-kinase and mediated by the NADPH-dependent phagocyte oxidase. J Biol Chem 276, 3548235493.CrossRefGoogle Scholar
15. Anand, K & Kaul, D (2003) Vitamin D3-dependent pathway regulates TACO gene transcription. Biochem Biophys Res Commun 310, 876877.CrossRefGoogle ScholarPubMed
16. Hmama, Z, Sendide, K, Talal, A et al. (2004) Quantitative analysis of phagolysosome fusion in intact cells: Inhibition by mycobacterial lipoarabinomannan and rescue by an 1alpha,25-dihydroxyvitamin D3-phosphoinositide 3-kinase pathway. J Cell Sci 117, 21312140.CrossRefGoogle Scholar
17. Coussens, A, Timms, PM, Boucher, BJ et al. (2009) 1alpha,25-dihydroxyvitamin D3 inhibits matrix metalloproteinases induced by Mycobacterium tuberculosis infection. Immunology 127, 539548.CrossRefGoogle Scholar
18. Liu, PT, Schenk, M, Walker, VP et al. (2009) Convergence of IL-1beta and VDR activation pathways in human TLR2/1-induced antimicrobial responses. PLoS ONE 4, e5810.CrossRefGoogle ScholarPubMed
19. Martineau, AR, Newton, SM, Wilkinson, KA et al. (2007) Neutrophil-mediated innate immune resistance to mycobacteria. J Clin Invest 117, 19881994.CrossRefGoogle ScholarPubMed
20. Yuk, JM, Shin, DM, Lee, HM et al. (2009) Vitamin D3 induces autophagy in human monocytes/macrophages via cathelicidin. Cell Host Microbe 6, 231243.CrossRefGoogle ScholarPubMed
21. Shin, DM, Yuk, JM, Lee, HM et al. (2010) Mycobacterial lipoprotein activates autophagy via TLR2/1/CD14 and a functional vitamin D receptor signalling. Cell Microbiol 12, 16481665.CrossRefGoogle Scholar
22. Liu, PT, Stenger, S, Tang, DH et al. (2007) Cutting edge: Vitamin D-mediated human antimicrobial activity against Mycobacterium tuberculosis is dependent on the induction of cathelicidin. J Immunol 179, 20602063.CrossRefGoogle ScholarPubMed
23. Glisson, F, Bate, G & Regemorter, A (1651) A Treatise of the Rickets: being A Disease Common to Children. London: P. Cole.Google Scholar
24. De Ugarte, DA, Shapiro, NL & Williams, HL (2003) Tuberculous mediastinal mass presenting with stridor in a 3-month-old child. J Pediatr Surg 38, 624625.CrossRefGoogle Scholar
25. Chapman, HT (1849) On the use of cod-liver oil in diseases of the bones and joints, in consumption and in other maladies attended by great emaciation. Pharm Trans, 116.Google Scholar
26. Karyadi, E, West, CE, Schultink, W et al. (2002) A double-blind, placebo-controlled study of vitamin A and zinc supplementation in persons with tuberculosis in Indonesia: Effects on clinical response and nutritional status. Am J Clin Nutr 75, 720727.CrossRefGoogle ScholarPubMed
27. Mayer, E (1938) Heliotherapy of tuberculosis. Ann Int Med 11, 18561860.Google Scholar
28. Roelandts, R (2005) A new light on Niels Finsen, a century after his Nobel Prize. Photodermatol Photoimmunol Photomed 21, 115117.CrossRefGoogle Scholar
29. Askew, FA, Bruce, HM, Callow, RK et al. (1931) Crystalline vitamin D. Nature 128, 758.CrossRefGoogle Scholar
30. Charpy, J (1950) Quelques traitments vitaminés ou par substances fonctionelles en dermatologie. Bull Méd 24, 505.Google Scholar
31. Martineau, AR, Honecker, FU, Wilkinson, RJ et al. (2007) Vitamin D in the treatment of pulmonary tuberculosis. J Steroid Biochem Mol Biol 103, 793798.CrossRefGoogle ScholarPubMed
32. Davies, PD (1985) A possible link between vitamin D deficiency and impaired host defence to Mycobacterium tuberculosis . Tubercle 66, 301306.CrossRefGoogle ScholarPubMed
33. Davies, PD, Brown, RC & Woodhead, JS (1985) Serum concentrations of vitamin D metabolites in untreated tuberculosis. Thorax 40, 187190.CrossRefGoogle ScholarPubMed
34. Davies, PD, Church, HA, Brown, RC et al. (1987) Raised serum calcium in tuberculosis patients in Africa. Eur J Respir Dis 71, 341344.Google ScholarPubMed
35. Davies, PD, Church, HA, Bovornkitti, S et al. (1988) Altered vitamin D homeostasis in tuberculosis. Int Med Thailand 4, 4547.Google Scholar
36. Wilkinson, RJ, Llewelyn, M, Toossi, Z et al. (2000) Influence of vitamin D deficiency and vitamin D receptor polymorphisms on tuberculosis among Gujarati Asians in west London: A case-control study. Lancet 355, 618621.CrossRefGoogle ScholarPubMed
37. Sasidharan, PK, Rajeev, E & Vijayakumari, V (2002) Tuberculosis and vitamin D deficiency. J Assoc Physicians India 50, 554558.Google ScholarPubMed
38. Sita-Lumsden, A, Lapthorn, G, Swaminathan, R et al. (2007) Reactivation of tuberculosis and vitamin D deficiency: The contribution of diet and exposure to sunlight. Thorax 62, 10031007.CrossRefGoogle ScholarPubMed
39. Gibney, KB, MacGregor, L, Leder, K et al. (2008) Vitamin D deficiency is associated with tuberculosis and latent tuberculosis infection in immigrants from sub-Saharan Africa. Clin Infect Dis 46, 443446.CrossRefGoogle ScholarPubMed
40. Ho-Pham, LT, Nguyen, ND, Nguyen, TT et al. (2010) Association between vitamin D insufficiency and tuberculosis in a Vietnamese population. BMC Infect Dis 10, 306.CrossRefGoogle Scholar
41. Grange, JM, Davies, PD, Brown, RC et al. (1985) A study of vitamin D levels in Indonesian patients with untreated pulmonary tuberculosis. Tubercle 66, 187191.CrossRefGoogle ScholarPubMed
42. Chan, TY, Poon, P, Pang, J et al. (1994) A study of calcium and vitamin D metabolism in Chinese patients with pulmonary tuberculosis. J Trop Med Hyg 97, 2630.Google ScholarPubMed
43. Martineau, AR, Leandro, AC, Anderson, ST et al. (2010) Association between Gc genotype and susceptibility to TB is dependent on vitamin D status. Eur Respir J 35, 11061112.CrossRefGoogle Scholar
44. Nielsen, NO, Skifte, T, Andersson, M et al. (2010) Both high and low serum vitamin D concentrations are associated with tuberculosis: A case-control study in Greenland. Br J Nutr 104, 14871489.CrossRefGoogle ScholarPubMed
45. Bellamy, R, Ruwende, C, Corrah, T et al. (1999) Tuberculosis and chronic hepatitis B virus infection in Africans and variation in the vitamin D receptor gene. J Infect Dis 179, 721724.CrossRefGoogle ScholarPubMed
46. Gao, L, Tao, Y, Zhang, L et al. (2010) Vitamin D receptor genetic polymorphisms and tuberculosis: Updated systematic review and meta-analysis. Int J Tuberc Lung Dis 14, 1523.Google ScholarPubMed
47. Arnaud, J & Constans, J (1993) Affinity differences for vitamin D metabolites associated with the genetic isoforms of the human serum carrier protein (DBP). Hum Genet 92, 183188.CrossRefGoogle ScholarPubMed
48. Lauridsen, AL, Vestergaard, P, Hermann, AP et al. (2005) Plasma concentrations of 25-hydroxy-vitamin D and 1,25-dihydroxy-vitamin D are related to the phenotype of Gc (vitamin D-binding protein): A cross-sectional study on 595 early postmenopausal women. Calcif Tissue Int 77, 1522.CrossRefGoogle Scholar
49. Abbas, S, Linseisen, J, Slanger, T et al. (2008) The Gc2 allele of the vitamin D binding protein is associated with a decreased postmenopausal breast cancer risk, independent of the vitamin D status. Cancer Epidemiol Biomarkers Prev 17, 13391343.CrossRefGoogle ScholarPubMed
50. Papiha, SS, Agarwal, SS & White, I (1983) Association between phosphoglucomutase (PGM1) and group-specific component (Gc) subtypes and tuberculosis. J Med Genet 20, 220222.CrossRefGoogle ScholarPubMed
51. Spitsyn, VA & Titenko, NV (1990) Subtypes of serum group specific component (Gc) in normal conditions and in pathology. Genetika 26, 749759.Google Scholar
52. Bahr, GM, Eales, LJ, Nye, KE et al. (1989) An association between Gc (vitamin D-binding protein) alleles and susceptibility to rheumatic fever. Immunology 67, 126128.Google ScholarPubMed
53. Roth, DE, Soto, G, Arenas, F et al. (2004) Association between vitamin D receptor gene polymorphisms and response to treatment of pulmonary tuberculosis. J Infect Dis 190, 920927.CrossRefGoogle ScholarPubMed
54. Babb, C, van der Merwe, L, Beyers, N et al. (2007) Vitamin D receptor gene polymorphisms and sputum conversion time in pulmonary tuberculosis patients. Tuberculosis (Edinb) 87, 295302.CrossRefGoogle ScholarPubMed
55. Talat, N, Perry, S, Parsonnet, J et al. (2010) Vitamin D deficiency and tuberculosis progression. Emerg Infect Dis 16, 853855.CrossRefGoogle ScholarPubMed
56. Kampmann, B, Gaora, PO, Snewin, VA et al. (2000) Evaluation of human antimycobacterial immunity using recombinant reporter mycobacteria. J Infect Dis 182, 895901.CrossRefGoogle ScholarPubMed
57. Martineau, AR, Wilkinson, RJ, Wilkinson, KA et al. (2007) A single dose of vitamin D enhances immunity to mycobacteria. Am J Respir Crit Care Med 176, 208213.CrossRefGoogle ScholarPubMed
58. Gwinup, G, Randazzo, G & Elias, A (1981) The influence of vitamin D intake on serum calcium in tuberculosis. Acta Endocrinol (Copenh) 97, 114117.Google ScholarPubMed
59. Martineau, AR, Nanzer, AM, Satkunam, KR et al. (2009) Influence of a single oral dose of vitamin D2 on serum 25-hydroxyvitamin D concentrations in tuberculosis patients. Int J Tuberc Lung Dis 13, 119125.Google ScholarPubMed
60. Narang, NK, Gupta, RC & Jain, MK (1984) Role of vitamin D in pulmonary tuberculosis. J Assoc Physicians India 32, 185188.Google ScholarPubMed
61. Tjellesen, L, Hummer, L, Christiansen, C et al. (1986) Serum concentration of vitamin D metabolites during treatment with vitamin D2 and D3 in normal premenopausal women. Bone Miner 1, 407413.Google ScholarPubMed
62. Stern, PH, Taylor, AB, Bell, NH et al. (1981) Demonstration that circulating 1 alpha, 25-dihydroxyvitamin D is loosely regulated in normal children. J Clin Invest 68, 13741377.CrossRefGoogle ScholarPubMed
63. Morcos, MM, Gabr, AA, Samuel, S et al. (1998) Vitamin D administration to tuberculous children and its value. Boll Chim Farm 137, 157164.Google ScholarPubMed
64. Nursyam, EW, Amin, Z & Rumende, CM (2006) The effect of vitamin D as supplementary treatment in patients with moderately advanced pulmonary tuberculous lesion. Acta Med Indones 38, 35.Google ScholarPubMed
65. Wejse, C, Gomes, VF, Rabna, P et al. (2009) Vitamin D as supplementary treatment for tuberculosis: A double-blind, randomized, placebo-controlled trial. Am J Respir Crit Care Med 179, 843850.CrossRefGoogle ScholarPubMed
66. Martineau, AR, Timms, PM, Bothamley, GH et al. (2011) High-dose vitamin D3 during intensive-phase antimicrobial treatment of pulmonary tuberculosis: A double-blind randomised controlled trial. Lancet 377, 242250.CrossRefGoogle ScholarPubMed
67. Vieth, R (2011) Vitamin D nutrient to treat TB begs the prevention question. Lancet 377, 189190.CrossRefGoogle ScholarPubMed
68. Ferebee, SH (1970) Controlled chemoprophylaxis trials in tuberculosis. A general review. Bibl Tuberc 26, 28–106.Google ScholarPubMed
Figure 0

Table 1. Summary of randomised controlled trials investigating effects of adjunctive vitamin D in patients with tuberculosis (TB)