Skip to main content

Advertisement

Log in

The role of UV radiation and vitamin D in the seasonality and outcomes of infectious disease

  • Perspective
  • Published:
Photochemical & Photobiological Sciences Aims and scope Submit manuscript

Abstract

The seasonality of infectious disease outbreaks suggests that environmental conditions have a significant effect on disease risk. One of the major environmental factors that can affect this is solar radiation, primarily acting through ultraviolet radiation (UVR), and its subsequent control of vitamin D production. Here we show how UVR and vitamin D, which are modified by latitude and season, can affect host and pathogen fitness and relate them to the outcomes of bacterial, viral and vector-borne infections. We conducted a thorough comparison of the molecular and cellular mechanisms of action of UVR and vitamin D on pathogen fitness and host immunity and related these to the effects observed in animal models and clinical trials to understand their independent and complementary effects on infectious disease outcome. UVR and vitamin D share common pathways of innate immune activation primarily via antimicrobial peptide production, and adaptive immune suppression. Whilst UVR can induce vitamin D-independent effects in the skin, such as the generation of photoproducts activating interferon signaling, vitamin D has a larger systemic effect due to its autocrine and paracrine modulation of cellular responses in a range of tissues. However, the seasonal patterns in infectious disease prevalence are not solely driven by variation in UVR and vitamin D levels across latitudes. Vector-borne pathogens show a strong seasonality of infection correlated to climatic conditions favoring their replication. Conversely, pathogens, such as influenza A virus, Mycobacterium tuberculosis and human immunodeficiency virus type 1, have strong evidence to support their interaction with vitamin D. Thus, UVR has both vitamin D-dependent and independent effects on infectious diseases; these effects vary depending on the pathogen of interest and the effects can be complementary or antagonistic.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

Abbreviations

1,25(OH)2D:

1α,25-Dihydroxy-vitamin D

25(OH)D:

25-Hydroxy-vitamin D

7-DHC:

7-Dehydrocholesterol

AHR:

Aryl hydrocarbon receptor

AMP:

Antimicrobial peptide

APC:

Antigen presenting cell

ARV:

Antiretroviral

BCG:

Bacillus Calmette-Guérin

CPD:

Cyclobutane pyrimidine dimers

dDC:

Dermal dendritic cell

dLN:

Draining lymph node

DC:

Dendritic cell

DTH:

Delayed type hypersensitivity

EBV:

Epstein-Barr virus

ECM:

Extracellular matrix

HIV:

Human immunodeficiency virus

HSV:

Herpes simplex virus type 1

IAV:

Influenza A virus

IL:

Interleukin

LC:

Langerhans cells

LPS:

Lipopolysaccharide

MAPK:

Mitogen-activated protein kinases

MC:

Mast Cell

MDM:

Monocyte-derived macrophages

MN:

Monocyte

mVDR:

Membrane vitamin D receptor

NK:

Natural Killer cells

NO:

Nitric oxide

nVDR:

Nuclear vitamin D receptor

PBMC:

Peripheral blood mononuclear cells

PI3K:

Phosphatidylinositol 3-kinase

PP:

Photo products

PRR:

Pattern recognition receptor

ROS:

Reactive oxygen species

RSV:

Respiratory syncytial virus (RSV)

RTI:

Respiratory tract infections

RXR:

Retinoid X receptor

TB:

Tuberculosis

Th:

T helper

TLR:

Toll-like receptors

Treg:

Regulatory T cells

UCA:

trans-Urocanic acid

UVA:

Ultraviolet A radiation

UVB:

Ultraviolet B radiation

UVR:

Ultraviolet radiation

VDRE:

Vitamin D response elements

Notes and references

  1. A. R. Martineau, S. Nhamoyebonde, T. Oni, M. X. Rangaka, S. Marais, N. Bangani, et al., Reciprocal seasonal variation in vitamin D status and tuberculosis notifications in Cape Town, South Africa, Proc. Natl. Acad. Sci. U. S. A., 2011, 108, 47 19013–19017

    Article  PubMed  PubMed Central  Google Scholar 

  2. WHO, Using climate to predict disease outbreaks: a review, World Health Organisation, Geneva, 2004.

    Google Scholar 

  3. D. N. Fisman, Seasonality of infectious diseases, Annu. Rev. Public Health, 2007, 28, 127–143

    Article  PubMed  Google Scholar 

  4. F. B. Mayr, S. Yende and D. C. Angus, Epidemiology of severe sepsis, Virulence, 2014, 5, 1 4–11

    Article  PubMed  Google Scholar 

  5. N. C. Grassly and C. Fraser, Seasonal infectious disease epidemiology, Proc. Biol. Sci., 2006, 273, 1600 2541–2550

    PubMed  PubMed Central  Google Scholar 

  6. K. A. Murray, N. Preston, T. Allen, C. Zambrana-Torrelio, P. R. Hosseini and P. Daszak, Global biogeography of human infectious diseases, Proc. Natl. Acad. Sci. U. S. A., 2015, 112, 41 12746–12751

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. V. Guernier, M. E. Hochberg and J. F. Guegan, Ecology drives the worldwide distribution of human diseases, PLoS Biol., 2004, 2, 6 e141

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. M. Grigalavicius, J. Moan, A. Dahlback and A. Juzeniene, Daily, seasonal, and latitudinal variations in solar ultraviolet A and B radiation in relation to vitamin D production and risk for skin cancer, Int. J. Dermatol., 2016, 55, 1 e23–e28

    Article  CAS  PubMed  Google Scholar 

  9. O. Engelsen, The relationship between ultraviolet radiation exposure and vitamin D status, Nutrients, 2010, 2, 5 482–495

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. B. Lehmann and M. Meurer, Vitamin D metabolism, Dermatol. Ther., 2010, 23, 1 2–12

    Article  PubMed  Google Scholar 

  11. A. T. Slominski, Z. Janjetovic, B. E. Fuller, M. A. Zmijewski, R. C. Tuckey, M. N. Nguyen, et al., Products of vitamin D3 or 7-dehydrocholesterol metabolism by cytochrome P450scc show anti-leukemia effects, having low or absent calcemic activity, PLoS One, 2010, 5, 3 e9907

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. W. E. Bradshaw and C. M. Holzapfel, Genetic shift in photoperiodic response correlated with global warming, Proc. Natl. Acad. Sci. U. S. A., 2001, 98, 25 14509–14511

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. E. S. Jensen, S. Lundbye-Christensen, L. Pedersen, H. T. Sorensen and H. C. Schonheyder, Seasonal variation in meningococcal disease in Denmark: relation to age and meningococcal phenotype, Scand. J. Infect. Dis., 2003, 35, 4 226–229

    PubMed  Google Scholar 

  14. B. Sultan, K. Labadi, J. F. Guegan and S. Janicot, Climate drives the meningitis epidemics onset in west Africa, PLoS Med., 2005, 2, 1 e6

    Article  PubMed  PubMed Central  Google Scholar 

  15. R. R. Colwell, Global climate and infectious disease: the cholera paradigm, Science, 1996, 274, 5295 2025–2031

    Article  CAS  PubMed  Google Scholar 

  16. R. Colwell, P. Epstein, D. Gubler, M. Hall, P. Reiter, J. Shukla, et al., Global climate change and infectious diseases, Emerg. Infect. Dis., 1998, 4, 3 451–452

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. A. T. Maurelli, A. E. Hromockyj and M. L. Bernardini, Environmental regulation of Shigella virulence, Curr. Top. Microbiol. Immunol., 1992, 180, 95–116

    CAS  PubMed  Google Scholar 

  18. D. A. Ridenour, S. L. Cirillo, S. Feng, M. M. Samrakandi and J. D. Cirillo, Identification of a gene that affects the efficiency of host cell infection by Legionella pneumophila in a temperature-dependent fashion, Infect. Immun., 2003, 71, 11 6256–6263

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. G. Kapperud, E. Namork and H. J. Skarpeid, Temperature-inducible surface fibrillae associated with the virulence plasmid of Yersinia enterocolitica and Yersinia pseudotuberculosis, Infect. Immun., 1985, 47, 2 561–566

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. H. Y. Kwon, S. W. Kim, M. H. Choi, A. D. Ogunniyi, J. C. Paton, S. H. Park, et al., Effect of heat shock and mutations in ClpL and ClpP on virulence gene expression in Streptococcus pneumoniae, Infect. Immun., 2003, 71, 7 3757–3765

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. R. R. Dunn, T. J. Davies, N. C. Harris and M. C. Gavin, Global drivers of human pathogen richness and prevalence, Proc. Biol. Sci., 2010, 277, 1694 2587–2595

    PubMed  PubMed Central  Google Scholar 

  22. S. F. Dowell, Seasonal variation in host susceptibility and cycles of certain infectious diseases, Emerg. Infect. Dis., 2001, 7, 3 369–374

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. J. J. Cannell, R. Vieth, J. C. Umhau, M. F. Holick, W. B. Grant, S. Madronich, et al., Epidemic influenza and vitamin D, Epidemiol. Infect., 2006, 134, 6 1129–1140

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. A. Juzeniene, L. W. Ma, M. Kwitniewski, G. A. Polev, Z. Lagunova, A. Dahlback, et al., The seasonality of pandemic and non-pandemic influenzas: the roles of solar radiation and vitamin D, Int. J. Infect. Dis., 2010, 14, 12 e1099–e1105

    Article  PubMed  Google Scholar 

  25. P. O. Lang and D. Samaras, Aging adults and seasonal influenza: does the vitamin d status (h)arm the body?, J. Aging Res., 2012, 2012, 806198

    Article  PubMed  Google Scholar 

  26. D. J. Berry, K. Hesketh, C. Power and E. Hypponen, Vitamin D status has a linear association with seasonal infections and lung function in British adults, Br. J. Nutr., 2011, 106, 9 1433–1440

    Article  CAS  PubMed  Google Scholar 

  27. B. A. Cunha, Influenza: historical aspects of epidemics and pandemics, Infect. Dis. Clin. North Am., 2004, 18, 1 141–155

    Article  PubMed  Google Scholar 

  28. E. Tognotti, Influenza pandemics: a historical retrospect, J. Infect. Dev. Ctries., 2009, 3, 5 331–334

    Article  PubMed  Google Scholar 

  29. F. T. Chew, S. Doraisingham, A. E. Ling, G. Kumarasinghe and B. W. Lee, Seasonal trends of viral respiratory tract infections in the tropics, Epidemiol. Infect., 1998, 121, 1 121–128

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. J. J. Cannell, M. Zasloff, C. F. Garland, R. Scragg and E. Giovannucci, On the epidemiology of influenza, Virol. J., 2008, 5, 29

    Article  PubMed  PubMed Central  Google Scholar 

  31. C. K. Li, B. C. Choi and T. W. Wong, Influenza-related deaths and hospitalizations in Hong Kong: a subtropical area, Public Health, 2006, 120, 6 517–524

    Article  CAS  PubMed  Google Scholar 

  32. J. Moan, A. Dahlback, L. Ma and A. Juzeniene, Influenza, solar radiation and vitamin D, Dermatoendocrinology, 2009, 1, 6 307–309

    Article  CAS  Google Scholar 

  33. A. S. Shadrin, I. G. Marinich and L. Y. Taros, Experimental and epidemiological estimation of seasonal and climato-geographical features of non-specific resistance of the organism to influenza, J. Hyg., Epidemiol., Microbiol., Immunol., 1977, 21, 2 155–161

    CAS  Google Scholar 

  34. M. P. Zykov and A. V. Sosunov, Vaccination activity of live influenza vaccine in different seasons of the year, J. Hyg., Epidemiol., Microbiol., Immunol., 1987, 31, 4 453–459

    CAS  Google Scholar 

  35. L. G. Stensballe, J. K. Devasundaram and E. A. Simoes, Respiratory syncytial virus epidemics: the ups and downs of a seasonal virus, Pediatr. Infect. Dis. J., 2003, 22, 2 Suppl S21–S32

    PubMed  Google Scholar 

  36. M. Koopmans and D. Brown, Seasonality and diversity of Group A rotaviruses in Europe, Acta Paediatr. Suppl., 1999, 88, 426 14–19

    Article  CAS  PubMed  Google Scholar 

  37. Y. Zhou, L. Li, B. Kim, K. Kaneshi, S. Nishimura, T. Kuroiwa, et al., Rotavirus infection in children in Japan, Pediatr. Int., 2000, 42, 4 428–439

    Article  CAS  PubMed  Google Scholar 

  38. S. M. Cook, R. I. Glass, C. W. LeBaron and M. S. Ho, Global seasonality of rotavirus infections, Bull. World Health Organ., 1990, 68, 2 171–177

    CAS  PubMed  PubMed Central  Google Scholar 

  39. F. Termorshuizen, R. B. Geskus, M. T. Roos, R. A. Coutinho, H. Van Loveren, Seasonal influences on immunological parameters in HIV-infected homosexual men: searching for the immunomodulating effects of sunlight, Int. J. Hyg. Environ. Health, 2002, 205, 5 379–384

    Article  PubMed  Google Scholar 

  40. J. H. Maclachlan, C. J. Lavender and B. C. Cowie, Effect of latitude on seasonality of tuberculosis, Australia, 2002–2011, Emerg. Infect. Dis., 2012, 18, 11 1879–1881

    Article  PubMed  PubMed Central  Google Scholar 

  41. L. E. Thorpe, T. R. Frieden, K. F. Laserson, C. Wells and G. R. Khatri, Seasonality of tuberculosis in India: is it real and what does it tell us?, Lancet, 2004, 364, 9445 1613–1614

    Article  PubMed  Google Scholar 

  42. M. D. Willis, C. A. Winston, C. M. Heilig, K. P. Cain, N. D. Walter, W. R. Mac Kenzie, Seasonality of tuberculosis in the United States, 1993–2008, Clin. Infect. Dis., 2012, 54, 11 1553–1560

    Article  PubMed  Google Scholar 

  43. R. J. Wilkinson, M. Llewelyn, Z. Toossi, P. Patel, G. Pasvol, A. Lalvani, et al., Influence of vitamin D deficiency and vitamin D receptor polymorphisms on tuberculosis among Gujarati Asians in west London: a case-control study, Lancet, 2000, 355, 9204 618–621

    Article  CAS  PubMed  Google Scholar 

  44. N. Talat, S. Perry, J. Parsonnet, G. Dawood and R. Hussain, Vitamin D deficiency and tuberculosis progression, Emerg. Infect. Dis., 2010, 16, 5 853–855

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. A. Fares, Seasonality of tuberculosis, J. Global Infect. Dis., 2011, 3, 1 46–55

    Article  Google Scholar 

  46. A. M. Molesworth, M. C. Thomson, S. J. Connor, M. P. Cresswell, A. P. Morse, P. Shears, et al., Where is the meningitis belt? Defining an area at risk of epidemic meningitis in Africa, Trans. R. Soc. Trop. Med. Hyg., 2002, 96, 3 242–249

    Article  PubMed  Google Scholar 

  47. A. J. Pollard and D. Scheifele, Meningococcal disease and vaccination in North America, J. Paediatr. Child Health, 2001, 37, 5 S20–S27

    Article  CAS  PubMed  Google Scholar 

  48. M. Emch, C. Feldacker, M. S. Islam and M. Ali, Seasonality of cholera from 1974 to 2005: a review of global patterns, Int. J. Health Geogr., 2008, 7, 31

    Article  PubMed  PubMed Central  Google Scholar 

  49. W. Checkley, L. D. Epstein, R. H. Gilman, D. Figueroa, R. I. Cama, J. A. Patz, et al., Effect of El Nino and ambient temperature on hospital admissions for diarrhoeal diseases in Peruvian children, Lancet, 2000, 355, 9202 442–450

    Article  CAS  PubMed  Google Scholar 

  50. M. S. Islam, B. S. Drasar and R. B. Sack, The aquatic flora and fauna as reservoirs of Vibrio cholerae: a review, J. Diarrhoeal. Dis. Res., 1994, 12, 2 87–96

    CAS  PubMed  Google Scholar 

  51. P. Siriyasatien, A. Phumee, P. Ongruk, K. Jampachaisri and K. Kesorn, Analysis of significant factors for dengue fever incidence prediction, BMC Bioinformatics, 2016, 17, 166

    Article  PubMed  PubMed Central  Google Scholar 

  52. E. Odongo-Aginya, G. Ssegwanyi, P. Kategere and P. C. Vuzi, Relationship between malaria infection intensity and rainfall pattern in Entebbe peninsula, Uganda, Afr. Health Sci., 2005, 5, 3 238–245

    CAS  PubMed  PubMed Central  Google Scholar 

  53. G. C. Ejezie and E. N. Ezedinachi, Malaria parasite density and body temperature in children under 10 years of age in Calabar, Nigeria, Trop. Geogr. Med., 1992, 44, 1–2 97–101

    CAS  PubMed  Google Scholar 

  54. P. Bouvier, A. Rougemont, N. Breslow, O. Doumbo, V. Delley, A. Dicko, et al., Seasonality and malaria in a west African village: does high parasite density predict fever incidence?, Am. J. Epidemiol., 1997, 145, 9 850–857

    Article  CAS  PubMed  Google Scholar 

  55. X. He, J. Yan, X. Zhu, Q. Wang, W. Pang, Z. Qi, et al., Vitamin D inhibits the occurrence of experimental cerebral malaria in mice by suppressing the host inflammatory response, J. Immunol., 2014, 193, 3 1314–1323

    Article  CAS  PubMed  Google Scholar 

  56. K. V. Luong and L. T. Nguyen, The role of vitamin D in malaria, J. Infect. Dev. Ctries., 2015, 9, 1 8–19

    Article  CAS  PubMed  Google Scholar 

  57. S. E. Cusick, R. O. Opoka, T. C. Lund, C. C. John and L. E. Polgreen, Vitamin D insufficiency is common in Ugandan children and is associated with severe malaria, PLoS One, 2014, 9, 12 e113185

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. A.-L. Khoo, H. J. P. M. Koenen, L. Y. A. Chai, F. C. G. J. Sweep, M. G. Netea, A. J. A. M. van der Ven, et al., Seasonal variation in vitamin D3 levels is paralleled by changes in the peripheral blood human T cell compartment, PLoS One, 2012, 7, 1 e29250

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. A. L. Khoo, L. Y. Chai, H. J. Koenen, F. C. Sweep, I. Joosten, M. G. Netea, et al., Regulation of cytokine responses by seasonality of vitamin D status in healthy individuals, Clin. Exp. Immunol., 2011, 164, 1 72–79

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. A. K. Coussens, C. E. Naude, R. Goliath, G. Chaplin, R. J. Wilkinson and N. G. Jablonski, High-dose vitamin D3 reduces deficiency caused by low UVB exposure and limits HIV-1 replication in urban Southern Africans, Proc. Natl. Acad. Sci. U. S. A., 2015, 112, 26 8052–8057

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. X. C. Dopico, M. Evangelou, R. C. Ferreira, H. Guo, M. L. Pekalski, D. J. Smyth, et al., Widespread seasonal gene expression reveals annual differences in human immunity and physiology, Nat. Commun., 2015, 6, 7000

    Article  CAS  PubMed  Google Scholar 

  62. T. J. John and P. Jayabal, Oral polio vaccination of children in the tropics. I. The poor seroconversion rates and the absence of viral interference, Am. J. Epidemiol., 1972, 96, 4 263–269

    Article  CAS  PubMed  Google Scholar 

  63. G. A. Colditz, T. F. Brewer, C. S. Berkey, M. E. Wilson, E. Burdick, H. V. Fineberg, et al., Efficacy of BCG vaccine in the prevention of tuberculosis. Meta-analysis of the published literature, J. Am. Med. Assoc., 1994, 271, 9 698–702

    Article  CAS  Google Scholar 

  64. T. A. Swartz, P. Skalska, C. G. Gerichter and W. C. Cockburn, Routine administration of oral polio vaccine in a subtropical area. Factors possibly influencing sero-conversion rates, J. Hyg., 1972, 70, 4 719–726

    CAS  PubMed  PubMed Central  Google Scholar 

  65. F. Termorshuizen, J. Garssen, M. Norval, L. Koulu, J. Laihia, L. Leino, et al., A review of studies on the effects of ultraviolet irradiation on the resistance to infections: evidence from rodent infection models and verification by experimental and observational human studies, Int. Immunopharmacol., 2002, 2, 2–3 263–275

    Article  CAS  PubMed  Google Scholar 

  66. N. Linder, Y. Abudi, W. Abdalla, M. Badir, Y. Amitai, J. Samuels, et al., Effect of season of inoculation on immune response to rubella vaccine in children, J. Trop. Pediatr., 2011, 57, 4 299–302

    Article  PubMed  Google Scholar 

  67. L. J. Rothschild, The influence of UV radiation on protistan evolution, J. Eukaryot. Microbiol., 1999, 46, 5 548–555

    Article  CAS  PubMed  Google Scholar 

  68. S. D. Keil, P. Kiser, J. J. Sullivan, A. S. Kong, H. L. Reddy, A. Avery, et al., Inactivation of Plasmodium spp. in plasma and platelet concentrates using riboflavin and ultraviolet light, Transfusion, 2013, 53, 10 2278–2286

    CAS  PubMed  Google Scholar 

  69. L. J. Cardo, F. J. Rentas, L. Ketchum, J. Salata, R. Harman, W. Melvin, et al., Pathogen inactivation of Leishmania donovani infantum in plasma and platelet concentrates using riboflavin and ultraviolet light, Vox Sang., 2006, 90, 2 85–91

    Article  CAS  PubMed  Google Scholar 

  70. A. P. Cap, H. F. Pidcoke, S. D. Keil, H. M. Staples, M. Anantpadma, R. Carrion Jr., et al., Treatment of blood with a pathogen reduction technology using ultraviolet light and riboflavin inactivates Ebola virus in vitro, Transfusion, 2016, 56, Suppl 1 S6–15

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. J. R. Wikler, N. Janssen, D. P. Bruynzeel and C. Nieboer, The effect of UV-light on pityrosporum yeasts: ultrastructural changes and inhibition of growth, Acta Derm. Venereol., 1990, 70, 1 69–71

    CAS  PubMed  Google Scholar 

  72. S. H. Silva, A. C. Guedes, B. Gontijo, A. M. Ramos, L. S. Carmo, L. M. Farias, et al., Influence of narrow-band UVB phototherapy on cutaneous microbiota of children with atopic dermatitis, J. Eur. Acad. Dermatol. Venereol., 2006, 20, 9 1114–1120

    Article  CAS  PubMed  Google Scholar 

  73. R. W. Truman and T. P. Gillis, The effect of ultraviolet light radiation on Mycobacterium leprae, Int. J. Lepr. Other Mycobact. Dis., 2000, 68, 1 11–17

    CAS  PubMed  Google Scholar 

  74. H. L. David, W. D. Jones Jr. and C. M. Newman, Ultraviolet light inactivation and photoreactivation in the mycobacteria, Infect. Immun., 1971, 4, 3 318–319

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. B. Zeina, J. Greenman, W. M. Purcell and B. Das, Killing of cutaneous microbial species by photodynamic therapy, Br. J. Dermatol., 2001, 144, 2 274–278

    Article  CAS  PubMed  Google Scholar 

  76. V. Patra, S. N. Byrne and P. Wolf, The Skin Microbiome: Is It Affected by UV-induced Immune Suppression?, Front. Microbiol., 2016, 7, 1235

    PubMed  PubMed Central  Google Scholar 

  77. S. E. Ullrich, Mechanisms underlying UV-induced immune suppression, Mutat Res., 2005, 571, 1–2 185–205

    Article  CAS  PubMed  Google Scholar 

  78. M. Norval, P. McLoone, A. Lesiak and J. Narbutt, The effect of chronic ultraviolet radiation on the human immune system, Photochem. Photobiol., 2008, 84, 1 19–28

    Article  CAS  PubMed  Google Scholar 

  79. B. E. Clausen and J. M. Kel, Langerhans cells: critical regulators of skin immunity?, Immunol. Cell Biol., 2010, 88, 4 351–360

    Article  CAS  PubMed  Google Scholar 

  80. G. G. Krueger and G. Stingl, Immunology/inflammation of the skin–a 50-year perspective, J. Invest. Dermatol., 1989, 92, 4 Suppl 32S–51S

    Article  CAS  PubMed  Google Scholar 

  81. P. S. Friedmann, I. Strickland, A. A. Memon and P. M. Johnson, Early time course of recruitment of immune surveillance in human skin after chemical provocation, Clin. Exp. Immunol., 1993, 91, 3 351–356

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. F. O. Nestle, P. Di Meglio, J. Z. Qin and B. J. Nickoloff, Skin immune sentinels in health and disease, Nat. Rev. Immunol., 2009, 9, 10 679–691

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. E. C. De Fabo and F. P. Noonan, Mechanism of immune suppression by ultraviolet irradiation in vivo. I. Evidence for the existence of a unique photoreceptor in skin and its role in photoimmunology, J. Exp. Med., 1983, 158, 1 84–98

    Article  PubMed  Google Scholar 

  84. R. L. Gallo and J. J. Bernard, Innate immune sensors stimulate inflammatory and immunosuppressive responses to UVB radiation, J. Invest. Dermatol., 2014, 134, 6 1508–1511

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. G. M. Halliday, S. N. Byrne, J. M. Kuchel, T. S. Poon and R. S. Barnetson, The suppression of immunity by ultraviolet radiation: UVA, nitric oxide and DNA damage, Photochem. Photobiol. Sci., 2004, 3, 8 736–740

    Article  CAS  PubMed  Google Scholar 

  86. P. H. Hart, S. Gorman, J. J. Finlay-Jones, Modulation of the immune system by UV radiation: more than just the effects of vitamin D?, Nat. Rev. Immunol., 2011, 11, 9 584–596

    Article  CAS  PubMed  Google Scholar 

  87. M. P. Stapelberg, R. B. Williams, S. N. Byrne and G. M. Halliday, The alternative complement pathway seems to be a UVA sensor that leads to systemic immunosuppression, J. Invest. Dermatol., 2009, 129, 11 2694–2701

    Article  CAS  PubMed  Google Scholar 

  88. P. J. Rochette, J. P. Therrien, R. Drouin, D. Perdiz, N. Bastien, E. A. Drobetsky, et al., UVA-induced cyclobutane pyrimidine dimers form predominantly at thymine-thymine dipyrimidines and correlate with the mutation spectrum in rodent cells, Nucleic Acids Res., 2003, 31, 11 2786–2794

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. M. G. Kemp, L. A. Lindsey-Boltz and A. Sancar, UV Light Potentiates STING (Stimulator of Interferon Genes)-dependent Innate Immune Signaling through Deregulation of ULK1 (Unc51-like Kinase 1), J. Biol. Chem., 2015, 290, 19 12184–12194

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. C. Esser, I. Bargen, H. Weighardt, T. Haarmann-Stemmann and J. Krutmann, Functions of the aryl hydrocarbon receptor in the skin, Semin. Immunopathol., 2013, 35, 6 677–691

    Article  CAS  PubMed  Google Scholar 

  91. Y. Xiang, G. Liu, L. Yang and J. L. Zhong, UVA-induced protection of skin through the induction of heme oxygenase-1, BioSci. Trends, 2011, 5, 6 239–244

    Article  CAS  PubMed  Google Scholar 

  92. L. Feldmeyer, M. Keller, G. Niklaus, D. Hohl, S. Werner and H. D. Beer, The inflammasome mediates UVB-induced activation and secretion of interleukin-1beta by keratinocytes, Curr. Biol., 2007, 17, 13 1140–1145

    Article  CAS  PubMed  Google Scholar 

  93. T. H. Nasti and L. Timares, Inflammasome activation of IL-1 family mediators in response to cutaneous photodamage, Photochem. Photobiol., 2012, 88, 5 1111–1125

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. J. Jans, G. A. Garinis, W. Schul, A. van Oudenaren, M. Moorhouse, M. Smid, et al., Differential role of basal keratinocytes in UV-induced immunosuppression and skin cancer, Mol. Cell. Biol., 2006, 26, 22 8515–8526

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. G. Soong, J. Chun, D. Parker and A. Prince, Staphylococcus aureus activation of caspase 1/calpain signaling mediates invasion through human keratinocytes, J. Infect. Dis., 2012, 205, 10 1571–1579

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. K. Kitur, S. Wachtel, A. Brown, M. Wickersham, F. Paulino, H. F. Penaloza, et al., Necroptosis Promotes Staphylococcus aureus Clearance by Inhibiting Excessive Inflammatory Signaling, Cell Rep., 2016, 16, 8 2219–2230

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Y. He, R. Fisher, S. Chowdhury, I. Sultana, C. P. Pereira, M. Bray, et al., Vaccinia virus induces rapid necrosis in keratinocytes by a STAT3-dependent mechanism, PLoS One, 2014, 9, 11 e113690

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. J. Schauber and R. L. Gallo, Antimicrobial peptides and the skin immune defense system, J. Allergy Clin. Immunol., 2009, 124, 3 Suppl 2 R13–R18

    Article  CAS  PubMed  Google Scholar 

  99. M. Frohm, B. Agerberth, G. Ahangari, M. Stahle-Backdahl, S. Liden, H. Wigzell, et al., The expression of the gene coding for the antibacterial peptide LL-37 is induced in human keratinocytes during inflammatory disorders, J. Biol. Chem., 1997, 272, 24 15258–15263

    Article  CAS  PubMed  Google Scholar 

  100. R. A. Dorschner, V. K. Pestonjamasp, S. Tamakuwala, T. Ohtake, J. Rudisill, V. Nizet, et al., Cutaneous injury induces the release of cathelicidin anti-microbial peptides active against group A Streptococcus, J. Invest. Dermatol., 2001, 117, 1 91–97

    Article  CAS  PubMed  Google Scholar 

  101. S. P. Hong, M. J. Kim, M. Y. Jung, H. Jeon, J. Goo, S. K. Ahn, et al., Biopositive effects of low-dose UVB on epidermis: coordinate upregulation of antimicrobial peptides and permeability barrier reinforcement, J. Invest. Dermatol., 2008, 128, 12 2880–2887

    Article  CAS  PubMed  Google Scholar 

  102. R. Glaser, F. Navid, W. Schuller, C. Jantschitsch, J. Harder, J. M. Schroder, et al., UV-B radiation induces the expression of antimicrobial peptides in human keratinocytes in vitro and in vivo, J. Allergy Clin. Immunol., 2009, 123, 5 1117–1123

    Article  CAS  PubMed  Google Scholar 

  103. F. Navid, M. Boniotto, C. Walker, K. Ahrens, E. Proksch, T. Sparwasser, et al., Induction of regulatory T cells by a murine beta-defensin, J. Immunol., 2012, 188, 2 735–743

    Article  CAS  PubMed  Google Scholar 

  104. T. Schwarz, The Many Faces of UVR-Induced Immunosuppression, J. Investig. Dermatol. Symp. Proc., 2015, 17, 1 22–23

    Article  PubMed  Google Scholar 

  105. J. Yu, N. Mookherjee, K. Wee, D. M. Bowdish, J. Pistolic, Y. Li, et al., Host defense peptide LL-37, in synergy with inflammatory mediator IL-1beta, augments immune responses by multiple pathways, J. Immunol., 2007, 179, 11 7684–7691

    Article  CAS  PubMed  Google Scholar 

  106. S. Salzer, S. Kresse, Y. Hirai, S. Koglin, M. Reinholz, T. Ruzicka, et al., Cathelicidin peptide LL-37 increases UVB-triggered inflammasome activation: possible implications for rosacea, J. Dermatol. Sci., 2014, 76, 3 173–179

    Article  CAS  PubMed  Google Scholar 

  107. N. K. Gibbs and M. Norval, Photoimmunosuppression: a brief overview, Photodermatol., Photoimmunol. Photomed., 2013, 29, 2 57–64

    Article  CAS  Google Scholar 

  108. E. Damiani and S. E. Ullrich, Understanding the connection between platelet-activating factor, a UV-induced lipid mediator of inflammation, immune suppression and skin cancer, Prog. Lipid Res., 2016, 63, 14–27

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. A. Sleijffers, J. Garssen, H. Van Loveren, Ultraviolet radiation, resistance to infectious diseases, and vaccination responses, Methods, 2002, 28, 1 111–121

    Article  CAS  PubMed  Google Scholar 

  110. A. Jeevan, R. Evans, E. L. Brown and M. L. Kripke, Effect of local ultraviolet irradiation on infections of mice with Candida albicans, Mycobacterium bovis BCG, and Schistosoma mansoni, J. Invest. Dermatol., 1992, 99, 1 59–64

    Article  CAS  PubMed  Google Scholar 

  111. A. Jeevan, S. E. Ullrich, M. De Gracia, R. Shah and Y. Sun, Mechanism of UVB-induced suppression of the immune response to Mycobacterium bovis bacillus Calmette-Guerin: role of cytokines on macrophage function, Photochem. Photobiol., 1996, 64, 2 259–266

    Article  CAS  PubMed  Google Scholar 

  112. A. Jeevan, K. Gilliam, H. Heard and M. L. Kripke, Effects of ultraviolet radiation on the pathogenesis of Mycobacterium lepraemurium infection in mice, Exp. Dermatol., 1992, 1, 3 152–160

    Article  CAS  PubMed  Google Scholar 

  113. J. Garssen, M. Norval, J. Crosby, P. Dortant, H. Van Loveren, The role of urocanic acid in UVB-induced suppression of immunity to Trichinella spiralis infection in the rat, Immunology, 1999, 96, 2 298–306

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. W. Goettsch, J. Garssen, A. de Klerk, T. M. Herremans, P. Dortant, F. R. de Gruijl, et al. Effects of ultraviolet-B exposure on the resistance to Listeria monocytogenes in the rat, Photochem. Photobiol., 1996, 63, 5 672–679

    Article  CAS  PubMed  Google Scholar 

  115. J. Garssen, H. Van der Vliet, A. De Klerk, W. Goettsch, J. A. Dormans, C. A. Bruggeman, et al., A rat cytomegalovirus infection model as a tool for immunotoxicity testing, Eur. J. Pharmacol., 1995, 292, 3–4 223–231

    CAS  PubMed  Google Scholar 

  116. K. Yamamoto, R. Ito, M. Koura and T. Kamiyama, UV-B irradiation increases susceptibility of mice to malarial infection, Infect. Immun., 2000, 68, 4 2353–2355

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Y. M. Denkins and M. L. Kripke, Effect of UV irradiation on lethal infection of mice with Candida albicans, Photochem. Photobiol., 1993, 57, 2 266–271

    Article  CAS  PubMed  Google Scholar 

  118. J. W. Gilmour and M. Norval, The effect of ultraviolet B irradiation, cis-urocanic acid and tumour necrosis factor-alpha on delayed hypersensitivity to herpes simplex virus, Photodermatol., Photoimmunol. Photomed., 1992, 9, 6 255–261

    CAS  Google Scholar 

  119. A. A. El-Ghorr, G. Horsburgh and M. Norval, The effect of UVB irradiation on antibody responses during herpes simplex virus type 1 (HSV-1) infections of mice, Photodermatol., Photoimmunol. Photomed., 1998, 14, 1 17–25

    Article  CAS  Google Scholar 

  120. L. K. Ryan, D. L. Neldon, L. R. Bishop, M. I. Gilmour, M. J. Daniels and D. M. Sailstad, et al. Exposure to ultraviolet radiation enhances mortality and pathology associated with influenza virus infection in mice, Photochem. Photobiol., 2000, 72, 4 497–507

    Article  CAS  PubMed  Google Scholar 

  121. E. L. Brown, S. E. Ullrich, M. Pride and M. L. Kripke, The effect of UV irradiation on infection of mice with Borrelia burgdorferi, Photochem. Photobiol., 2001, 73, 5 537–544

    Article  CAS  PubMed  Google Scholar 

  122. S. H. Giannini, Effects of ultraviolet B irradiation on cutaneous leishmaniasis, Parasitol. Today, 1992, 8, 2 44–48

    Article  CAS  PubMed  Google Scholar 

  123. M. D. Ward, D. M. Sailstad, D. L. Andrews, E. H. Boykin and M. K. Selgrade, Ultraviolet radiation downregulates allergy in BALB/c mice, J. Toxicol. Environ. Health, Part A, 2004, 67, 1 73–85

    Article  CAS  Google Scholar 

  124. N. L. Letvin, R. S. Kauffman and R. Finberg, T lymphocyte immunity to reovirus: cellular requirements for generation and role in clearance of primary infections, J. Immunol., 1981, 127, 6 2334–2339

    CAS  PubMed  Google Scholar 

  125. C. M. Brozek, G. M. Shopp, S. L. Ryan, P. M. Gillespie, D. F. Kusewitt, M. S. Rajagopalan, et al., In vivo exposure to ultraviolet radiation enhances pathogenic effects of murine leukemia virus, LP-BM5, in murine acquired immunodeficiency syndrome, Photochem. Photobiol., 1992, 56, 3 287–295

    Article  CAS  PubMed  Google Scholar 

  126. F. P. Noonan and F. A. Lewis, UVB-induced immune suppression and infection with Schistosoma mansoni, Photochem. Photobiol., 1995, 61, 1 99–105

    Article  CAS  PubMed  Google Scholar 

  127. M. Norval and G. M. Halliday, The consequences of UV-induced immunosuppression for human health, Photochem. Photobiol., 2011, 87, 5 965–977

    Article  CAS  PubMed  Google Scholar 

  128. M. Ichihashi, H. Nagai and K. Matsunaga, Sunlight is an important causative factor of recurrent herpes simplex, Cutis, 2004, 74, 5 Suppl 14–18

    PubMed  Google Scholar 

  129. D. D. Moyal and A. M. Fourtanier, Broad-spectrum sunscreens provide better protection from solar ultraviolet-simulated radiation and natural sunlight-induced immunosuppression in human beings, J. Am. Acad. Dermatol., 2008, 58, 5 Suppl 2 S149–S154

    Article  PubMed  Google Scholar 

  130. B. Guo, S. Naish, W. Hu and S. Tong, The potential impact of climate change and ultraviolet radiation on vaccine-preventable infectious diseases and immunization service delivery system, Expert Rev. Vaccines, 2015, 14, 4 561–577

    Article  CAS  PubMed  Google Scholar 

  131. M. K. Sharma, V. Bhatia and H. M. Swami, Outbreak of measles amongst vaccinated children in a slum of Chandigarh, Indian J. Med. Sci., 2004, 58, 2 47–53

    PubMed  Google Scholar 

  132. S. A. Snopov, S. M. Kharit, M. Norval and V. V. Ivanova, Circulating leukocyte and cytokine responses to measles and poliovirus vaccination in children after ultraviolet radiation exposures, Arch. Virol., 2005, 150, 9 1729–1743

    Article  CAS  PubMed  Google Scholar 

  133. A. Sleijffers, B. Yucesoy, M. Kashon, J. Garssen, F. R. De Gruijl, G. J. Boland, et al., Cytokine polymorphisms play a role in susceptibility to ultraviolet B-induced modulation of immune responses after hepatitis B vaccination, J. Immunol., 2003, 170, 6 3423–3428

    Article  CAS  PubMed  Google Scholar 

  134. A. Sleijffers, A. Kammeyer, F. R. de Gruijl, G. J. Boland, J. van Hattum, W. A. van Vloten, et al., Epidermal cis-urocanic acid levels correlate with lower specific cellular immune responses after hepatitis B vaccination of ultraviolet B-exposed humans, Photochem. Photobiol., 2003, 77, 3 271–275

    Article  CAS  PubMed  Google Scholar 

  135. M. C. Naranjo, J. M. Guerrero, A. Rubio, P. J. Lardone, A. Carrillo-Vico, M. P. Carrascosa-Salmoral, et al., Melatonin biosynthesis in the thymus of humans and rats, Cell. Mol. Life Sci., 2007, 64, 6 781–790

    Article  CAS  PubMed  Google Scholar 

  136. A. Carrillo-Vico, J. R. Calvo, P. Abreu, P. J. Lardone, S. Garcia-Maurino, R. J. Reiter, et al., Evidence of melatonin synthesis by human lymphocytes and its physiological significance: possible role as intracrine, autocrine, and/or paracrine substance, FASEB J., 2004, 18, 3 537–539

    Article  CAS  PubMed  Google Scholar 

  137. V. Srinivasan, D. W. Spence, I. Trakht, S. R. Pandi-Perumal, D. P. Cardinali and G. J. Maestroni, Immunomodulation by melatonin: its significance for seasonally occurring diseases, Neuroimmunomodulation, 2008, 15, 2 93–101

    Article  CAS  PubMed  Google Scholar 

  138. R. Hardeland, Melatonin in aging and disease -multiple consequences of reduced secretion, options and limits of treatment, Aging Dis., 2012, 3, 2 194–225

    PubMed  Google Scholar 

  139. K. Honma, S. Honma, M. Kohsaka and N. Fukuda, Seasonal variation in the human circadian rhythm: dissociation between sleep and temperature rhythm, Am. J. Physiol., 1992, 262, 5 Pt 2 R885–R891

    CAS  PubMed  Google Scholar 

  140. T. Ueno-Towatari, K. Norimatsu, K. Blazejczyk, H. Tokura and T. Morita, Seasonal variations of melatonin secretion in young females under natural and artificial light conditions in Fukuoka, Japan, J. Physiol. Anthropol., 2007, 26, 2 209–215

    Article  PubMed  Google Scholar 

  141. T. W. Fischer, T. W. Sweatman, I. Semak, R. M. Sayre, J. Wortsman and A. Slominski, Constitutive and UV-induced metabolism of melatonin in keratinocytes and cell-free systems, FASEB J., 2006, 20, 9 1564–1566

    Article  CAS  PubMed  Google Scholar 

  142. M. J. Barjavel, Z. Mamdouh, N. Raghbate and O. Bakouche, Differential expression of the melatonin receptor in human monocytes, J. Immunol., 1998, 160, 3 1191–1197

    CAS  PubMed  Google Scholar 

  143. M. A. Lopez-Gonzalez, J. R. Calvo, J. J. Segura and J. M. Guerrero, Characterization of melatonin binding sites in human peripheral blood neutrophils, Biotechnol. Ther., 1993, 4, 3–4 253–262

    CAS  PubMed  Google Scholar 

  144. D. L. Drazen and R. J. Nelson, Melatonin receptor subtype MT2 (Mel 1b) and not mt1 (Mel 1a) is associated with melatonin-induced enhancement of cell-mediated and humoral immunity, Neuroendocrinology, 2001, 74, 3 178–184

    Article  CAS  PubMed  Google Scholar 

  145. A. Carrillo-Vico, R. J. Reiter, P. J. Lardone, J. L. Herrera, R. Fernandez-Montesinos, J. M. Guerrero, et al., The modulatory role of melatonin on immune responsiveness, Curr. Opin. Investig. Drugs, 2006, 7, 5 423–431

    CAS  PubMed  Google Scholar 

  146. N. E. Waly and R. Hallworth, Circadian Pattern of Melatonin MT1 and MT2 Receptor Localization in the Rat Suprachiasmatic Nucleus, J. Circadian Rhythms, 2015, 13, 1

    Article  PubMed  PubMed Central  Google Scholar 

  147. S. Garcia-Maurino, D. Pozo, J. R. Calvo and J. M. Guerrero, Correlation between nuclear melatonin receptor expression and enhanced cytokine production in human lymphocytic and monocytic cell lines, J. Pineal Res., 2000, 29, 3 129–137

    Article  CAS  PubMed  Google Scholar 

  148. C. Pena, J. Rincon, A. Pedreanez, N. Viera and J. Mosquera, Chemotactic effect of melatonin on leukocytes, J. Pineal Res., 2007, 43, 3 263–269

    Article  CAS  PubMed  Google Scholar 

  149. C. R. Sudfeld, E. L. Giovannucci, S. Isanaka, S. Aboud, F. M. Mugusi, M. Wang, et al., Vitamin D Status and Incidence of Pulmonary Tuberculosis, Opportunistic Infections, and Wasting Among HIV-Infected Tanzanian Adults Initiating Antiretroviral Therapy, J. Infect. Dis., 2013, 207, 3 378–385

    Article  CAS  PubMed  Google Scholar 

  150. J. Charan, J. P. Goyal, D. Saxena and P. Yadav, Vitamin D for prevention of respiratory tract infections: A systematic review and meta-analysis, J. Pharmacol. Pharmacother., 2012, 3, 4 300–303

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. F. P. Havers, B. Detrick, S. W. Cardoso, S. Berendes, J. R. Lama, P. Sugandhavesa, et al., Change in vitamin d levels occurs early after antiretroviral therapy initiation and depends on treatment regimen in resource-limited settings, PLoS One, 2014, 9, 4 e95164

    Article  PubMed  PubMed Central  Google Scholar 

  152. A. Moodley, M. Qin, K. K. Singh and S. A. Spector, Vitamin D-related host genetic variants alter HIV disease progression in children, Pediatr. Infect. Dis. J., 2013, 32, 11 1230–1236

    Article  PubMed  Google Scholar 

  153. R. Janssen, L. Bont, C. L. Siezen, H. M. Hodemaekers, M. J. Ermers, G. Doornbos, et al., Genetic susceptibility to respiratory syncytial virus bronchiolitis is predominantly associated with innate immune genes, J. Infect. Dis., 2007, 196, 6 826–834

    Article  CAS  PubMed  Google Scholar 

  154. A. R. Martineau, A. C. Leandro, S. T. Anderson, S. M. Newton, K. A. Wilkinson, M. P. Nicol, et al., Association between Gc genotype and susceptibility to TB is dependent on vitamin D status, Eur. Respir. J., 2010, 35, 5 1106–1112

    Article  CAS  PubMed  Google Scholar 

  155. R. Bellamy, C. Ruwende, T. Corrah, K. P. McAdam, M. Thursz, H. C. Whittle, et al., Tuberculosis and chronic hepatitis B virus infection in Africans and variation in the vitamin D receptor gene, J. Infect. Dis., 1999, 179, 3 721–724

    Article  CAS  PubMed  Google Scholar 

  156. H. Loke, D. Bethell, C. X. Phuong, N. Day, N. White, J. Farrar, et al., Susceptibility to dengue hemorrhagic fever in vietnam: evidence of an association with variation in the vitamin d receptor and Fc gamma receptor IIa genes, Am. J. Trop. Med. Hyg., 2002, 67, 1 102–106

    Article  CAS  PubMed  Google Scholar 

  157. C. R. Sudfeld, E. L. Giovannucci, S. Isanaka, S. Aboud, F. M. Mugusi, M. Wang, et al., Vitamin D status and incidence of pulmonary tuberculosis, opportunistic infections, and wasting among HIV-infected Tanzanian adults initiating antiretroviral therapy, J. Infect. Dis., 2013, 207, 3 378–385

    Article  CAS  PubMed  Google Scholar 

  158. C. R. Sudfeld, M. Wang, S. Aboud, E. L. Giovannucci, F. M. Mugusi and W. W. Fawzi, Vitamin D and HIV progression among Tanzanian adults initiating antiretroviral therapy, PLoS One, 2012, 7, 6 e40036

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. J. N. Jarvis, T. Bicanic, A. Loyse, G. Meintjes, L. Hogan, C. H. Roberts, et al., Very low levels of 25-hydroxyvitamin D are not associated with immunologic changes or clinical outcome in South African patients with HIV-associated cryptococcal meningitis, Clin. Infect. Dis., 2014, 59, 4 493–500

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. M. Norval, A. K. Coussens, R. J. Wilkinson, L. Bornman, R. M. Lucas and C. Y. Wright, Vitamin D Status and Its Consequences for Health in South Africa, Int. J. Environ. Res. Public Health, 2016, 13, 10 1019

    Article  PubMed Central  CAS  Google Scholar 

  161. P. T. Liu, S. Stenger, H. Li, L. Wenzel, B. H. Tan, S. R. Krutzik, et al., Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response, Science, 2006, 311, 5768 1770–1773

    Article  CAS  PubMed  Google Scholar 

  162. M. R. Pinzone, M. Di Rosa, B. M. Celesia, F. Condorelli, M. Malaguarnera, G. Madeddu, et al., LPS and HIV gp120 modulate monocyte/macrophage CYP27B1 and CYP24A1 expression leading to vitamin D consumption and hypovitaminosis D in HIV-infected individuals, Eur. Rev. Med. Pharmacol. Sci., 2013, 17, 14 1938–1950

    CAS  PubMed  Google Scholar 

  163. J. E. Smith and D. S. Goodman, The turnover and transport of vitamin D and of a polar metabolite with the properties of 25-hydroxycholecalciferol in human plasma, J. Clin. Invest., 1971, 50, 10 2159–2167

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. B. S. Levine, F. R. Singer, G. F. Bryce, J. P. Mallon, O. N. Miller and J. W. Coburn, Pharmacokinetics and biologic effects of calcitriol in normal humans, J. Lab. Clin. Med., 1985, 105, 2 239–246

    CAS  PubMed  Google Scholar 

  165. C. A. Coughlan, S. H. Chotirmall, J. Renwick, T. Hassan, T. B. Low, G. Bergsson, et al., The effect of Aspergillus fumigatus infection on vitamin D receptor expression in cystic fibrosis, Am. J. Respir. Crit. Care Med., 2012, 186, 10 999–1007

    Article  CAS  PubMed  Google Scholar 

  166. S. P. Yenamandra, A. Lundin, V. Arulampalam, M. Yurchenko, S. Pettersson, G. Klein, et al., Expression profile of nuclear receptors upon Epstein–Barr virus induced B cell transformation, Exp. Oncol., 2009, 31, 2 92–96

    CAS  PubMed  Google Scholar 

  167. J. T. Go, S. E. Belisle, N. Tchitchek, T. M. Tumpey, W. Ma, J. A. Richt, et al., 2009 pandemic H1N1 influenza virus elicits similar clinical course but differential host transcriptional response in mouse, macaque, and swine infection models, BMC Genomics, 2012, 13, 627

    Article  PubMed  PubMed Central  Google Scholar 

  168. S. Ray, K. S. Kamath, R. Srivastava, D. Raghu, K. Gollapalli, R. Jain, et al., Serum proteome analysis of vivax malaria: An insight into the disease pathogenesis and host immune response, J. Proteomics, 2012, 75, 10 3063–3080

    Article  CAS  PubMed  Google Scholar 

  169. T. T. Wang, L. E. Tavera-Mendoza, D. Laperriere, E. Libby, N. B. MacLeod, Y. Nagai, et al., Large-scale in silico and microarray-based identification of direct 1,25-dihydroxyvitamin D3 target genes, Mol. Endocrinol., 2005, 19, 11 2685–2695

    Article  CAS  PubMed  Google Scholar 

  170. A. W. Norman, H. L. Henry, J. E. Bishop, X. D. Song, C. Bula and W. H. Okamura, Different shapes of the steroid hormone 1alpha,25(OH)(2)-vitamin D(3) act as agonists for two different receptors in the vitamin D endocrine system to mediate genomic and rapid responses, Steroids, 2001, 66, 3–5 147–158

    Article  CAS  PubMed  Google Scholar 

  171. S. Alvarez-Diaz, N. Valle, G. Ferrer-Mayorga, L. Lombardia, M. Herrera, O. Dominguez, et al., MicroRNA-22 is induced by vitamin D and contributes to its antiproliferative, antimigratory and gene regulatory effects in colon cancer cells, Hum. Mol. Genet., 2012, 21, 10 2157–2165

    Article  CAS  PubMed  Google Scholar 

  172. S. Chen, G. P. Sims, X. X. Chen, Y. Y. Gu, S. Chen and P. E. Lipsky, Modulatory effects of 1,25-dihydroxyvitamin D3 on human B cell differentiation, J. Immunol., 2007, 179, 3 1634–1647

    Article  CAS  PubMed  Google Scholar 

  173. J. Fritsche, K. Mondal, A. Ehrnsperger, R. Andreesen and M. Kreutz, Regulation of 25-hydroxyvitamin D3-1 alpha-hydroxylase and production of 1 alpha,25-dihydroxyvitamin D3 by human dendritic cells, Blood, 2003, 102, 9 3314–3316

    Article  CAS  PubMed  Google Scholar 

  174. H. Sigmundsdottir, J. Pan, G. F. Debes, C. Alt, A. Habtezion, D. Soler, et al., DCs metabolize sunlight-induced vitamin D3 to ‘program’ T cell attraction to the epidermal chemokine CCL27, Nat. Immunol., 2007, 8, 3 285–293

    Article  CAS  PubMed  Google Scholar 

  175. A. M. Ramirez, C. Wongtrakool, T. Welch, A. Steinmeyer, U. Zugel and J. Roman, Vitamin D inhibition of pro-fibrotic effects of transforming growth factor beta1 in lung fibroblasts and epithelial cells, J. Steroid Biochem. Mol. Biol., 2010, 118, 3 142–150

    Article  CAS  PubMed  Google Scholar 

  176. M. Shalita-Chesner, R. Koren, Y. A. Mekori, D. Baram, C. Rotem, U. A. Liberman, et al., 1,25-Dihydroxyvitamin D3 enhances degranulation of mast cells, Mol. Cell. Endocrinol., 1998, 142, 1–2 49–55

    Article  CAS  PubMed  Google Scholar 

  177. E. Gottfried, M. Rehli, J. Hahn, E. Holler, R. Andreesen and M. Kreutz, Monocyte-derived cells express CYP27A1 and convert vitamin D3 into its active metabolite, Biochem. Biophys. Res. Commun., 2006, 349, 1 209–213

    Article  CAS  PubMed  Google Scholar 

  178. L. M. Sly, M. Lopez, W. M. Nauseef and N. E. Reiner, 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., 2001, 276, 38 35482–35493

    Article  CAS  PubMed  Google Scholar 

  179. K. A. Rockett, R. Brookes, I. Udalova, V. Vidal, A. V. Hill and D. Kwiatkowski, 1,25-Dihydroxyvitamin D3 induces nitric oxide synthase and suppresses growth of Mycobacterium tuberculosis in a human macrophage-like cell line, Infect. Immun., 1998, 66, 11 5314–5321

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. R. A. Fratti, J. M. Backer, J. Gruenberg, S. Corvera and V. Deretic, Role of phosphatidylinositol 3-kinase and Rab5 effectors in phagosomal biogenesis and mycobacterial phagosome maturation arrest, J. Cell Biol., 2001, 154, 3 631–644

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. A. R. Martineau, K. A. Wilkinson, S. M. Newton, R. A. Floto, A. W. Norman, K. Skolimowska, et al., IFN-gamma- and TNF-Independent Vitamin D-Inducible Human Suppression of Mycobacteria: The Role of Cathelicidin LL-37, J. Immunol., 2007, 178, 11 7190–7198

    Article  CAS  PubMed  Google Scholar 

  182. A. F. Gombart, N. Borregaard and H. P. Koeffler, Human cathelicidin antimicrobial peptide (CAMP) gene is a direct target of the vitamin D receptor and is strongly up-regulated in myeloid cells by 1,25-dihydroxyvitamin D3, FASEB J., 2005, 19, 9 1067–1077

    Article  CAS  PubMed  Google Scholar 

  183. O. E. Sorensen, P. Follin, A. H. Johnsen, J. Calafat, G. S. Tjabringa, P. S. Hiemstra, et al., Human cathelicidin, hCAP-18, is processed to the antimicrobial peptide LL-37 by extracellular cleavage with proteinase 3, Blood, 2001, 97, 12 3951–3959

    Article  CAS  PubMed  Google Scholar 

  184. A. R. Martineau, S. M. Newton, K. A. Wilkinson, B. Kampmann, B. M. Hall, N. Nawroly, et al., Neutrophil-mediated innate immune resistance to mycobacteria, J. Clin. Invest., 2007, 117, 7 1988–1994

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. G. Diamond, N. Beckloff and L. K. Ryan, Host defense peptides in the oral cavity and the lung: similarities and differences, J. Dent. Res., 2008, 87, 10 915–927

    Article  CAS  PubMed  Google Scholar 

  186. F. Neville, M. Cahuzac, O. Konovalov, Y. Ishitsuka, K. Y. Lee, I. Kuzmenko, et al., Lipid headgroup discrimination by antimicrobial peptide LL-37: insight into mechanism of action, Biophys. J., 2006, 90, 4 1275–1287

    Article  CAS  PubMed  Google Scholar 

  187. C. B. Buck, P. M. Day, C. D. Thompson, J. Lubkowski, W. Lu, D. R. Lowy, et al., Human alpha-defensins block papillomavirus infection, Proc. Natl. Acad. Sci. U. S. A., 2006, 103, 5 1516–1521

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. M. D. Howell, J. F. Jones, K. O. Kisich, J. E. Streib, R. L. Gallo and D. Y. Leung, Selective killing of vaccinia virus by LL-37: implications for eczema vaccinatum, J. Immunol., 2004, 172, 3 1763–1767

    Article  CAS  PubMed  Google Scholar 

  189. P. G. Barlow, P. Svoboda, A. Mackellar, A. A. Nash, I. A. York, J. Pohl, et al., Antiviral activity and increased host defense against influenza infection elicited by the human cathelicidin LL-37, PLoS One, 2011, 6, 10 e25333

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. S. M. Currie, E. Gwyer Findlay, A. J. McFarlane, P. M. Fitch, B. Bottcher, N. Colegrave, et al., Cathelicidins Have Direct Antiviral Activity against Respiratory Syncytial Virus In Vitro and Protective Function In Vivo in Mice and Humans, J. Immunol., 2016, 196, 6 2699–2710

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. S. Hansdottir, M. M. Monick, S. L. Hinde, N. Lovan, D. C. Look and G. W. Hunninghake, Respiratory epithelial cells convert inactive vitamin D to its active form: potential effects on host defense, J. Immunol., 2008, 181, 10 7090–7099

    Article  CAS  PubMed  Google Scholar 

  192. A. Di Nardo, A. Vitiello and R. L. Gallo, Cutting edge: mast cell antimicrobial activity is mediated by expression of cathelicidin antimicrobial peptide, J. Immunol., 2003, 170, 5 2274–2278

    Article  CAS  PubMed  Google Scholar 

  193. B. Agerberth, J. Charo, J. Werr, B. Olsson, F. Idali, L. Lindbom, et al., The human antimicrobial and chemotactic peptides LL-37 and α-defensins are expressed by specific lymphocyte and monocyte populations, Blood, 2000, 96, 3086–3093

    Article  CAS  PubMed  Google Scholar 

  194. J. M. Yuk, D. M. Shin, H. M. Lee, C. S. Yang, H. S. Jin, K. K. Kim, et al., Vitamin D3 induces autophagy in human monocytes/macrophages via cathelicidin, Cell Host Microbe, 2009, 6, 3 231–243

    Article  CAS  PubMed  Google Scholar 

  195. P. D. Hart, M. R. Young, A. H. Gordon and K. H. Sullivan, Inhibition of phagosome-lysosome fusion in macrophages by certain mycobacteria can be explained by inhibition of lysosomal movements observed after phagocytosis, J. Exp. Med., 1987, 166, 4 933–946

    Article  CAS  PubMed  Google Scholar 

  196. C. Munz, Enhancing immunity through autophagy, Annu. Rev. Immunol., 2009, 27, 423–449

    Article  CAS  PubMed  Google Scholar 

  197. J. Sun, VDR/vitamin D receptor regulates autophagic activity through ATG16L1, Autophagy, 2016, 12, 6 1057–1058

    Article  CAS  PubMed  Google Scholar 

  198. G. B. Kyei, C. Dinkins, A. S. Davis, E. Roberts, S. B. Singh, C. Dong, et al., Autophagy pathway intersects with HIV-1 biosynthesis and regulates viral yields in macrophages, J. Cell Biol., 2009, 186, 2 255–268

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. G. R. Campbell and S. A. Spector, Hormonally active vitamin D3 (1alpha,25-dihydroxycholecalciferol) triggers autophagy in human macrophages that inhibits HIV-1 infection, J. Biol. Chem., 2011, 286, 21 18890–18902

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. M. Fabri, S. Stenger, D. M. Shin, J. M. Yuk, P. T. Liu, S. Realegeno, et al., Vitamin D is required for IFN-gamma-mediated antimicrobial activity of human macrophages, Sci. Transl. Med., 2011, 3, 104 104ra102

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  201. T. T. Wang, B. Dabbas, D. Laperriere, A. J. Bitton, H. Soualhine, L. E. Tavera-Mendoza, et al., Direct and indirect induction by 1,25-dihydroxyvitamin D3 of the NOD2/CARD15-defensin beta2 innate immune pathway defective in Crohn disease, J. Biol. Chem., 2010, 285, 4 2227–2231

    Article  CAS  PubMed  Google Scholar 

  202. P. T. Liu, M. Schenk, V. P. Walker, P. W. Dempsey, M. Kanchanapoomi, M. Wheelwright, et al., Convergence of IL-1beta and VDR activation pathways in human TLR2/1-induced antimicrobial responses, PLoS One, 2009, 4, 6 e5810

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  203. S. Kota, A. Sabbah, T. H. Chang, R. Harnack, Y. Xiang, X. Meng, et al., Role of human beta-defensin-2 during tumor necrosis factor-alpha/NF-kappaB-mediated innate antiviral response against human respiratory syncytial virus, J. Biol. Chem., 2008, 283, 33 22417–22429

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. M. E. Quinones-Mateu, M. M. Lederman, Z. Feng, B. Chakraborty, J. Weber, H. R. Rangel, et al., Human epithelial beta-defensins 2 and 3 inhibit HIV-1 replication, AIDS, 2003, 17, 16 F39–F48

    Article  CAS  PubMed  Google Scholar 

  205. Z. Feng, G. R. Dubyak, M. M. Lederman and A. Weinberg, Cutting edge: human beta defensin 3–a novel antagonist of the HIV-1 coreceptor CXCR4, J. Immunol., 2006, 177, 2 782–786

    Article  CAS  PubMed  Google Scholar 

  206. E. Hazrati, B. Galen, W. Lu, W. Wang, Y. Ouyang, M. J. Keller, et al., Human alpha- and beta-defensins block multiple steps in herpes simplex virus infection, J. Immunol., 2006, 177, 12 8658–8666

    Article  CAS  PubMed  Google Scholar 

  207. T. Tecle, M. R. White, D. Gantz, E. C. Crouch and K. L. Hartshorn, Human neutrophil defensins increase neutrophil uptake of influenza A virus and bacteria and modify virus-induced respiratory burst responses, J. Immunol., 2007, 178, 12 8046–8052

    Article  CAS  PubMed  Google Scholar 

  208. S. Ranjbar, L. D. Jasenosky, N. Chow and A. E. Goldfeld, Regulation of Mycobacterium tuberculosis-dependent HIV-1 transcription reveals a new role for NFAT5 in the toll-like receptor pathway, PLoS Pathog., 2012, 8, 4 e1002620

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. A. A. Giangreco and L. Nonn, The sum of many small changes: microRNAs are specifically and potentially globally altered by vitamin D3 metabolites, J. Steroid Biochem. Mol. Biol., 2013, 136, 86–93

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Y. Zhang, D. Y. Leung, B. N. Richers, Y. Liu, L. K. Remigio, D. W. Riches, et al., Vitamin D inhibits monocyte/macrophage proinflammatory cytokine production by targeting MAPK phosphatase-1, J. Immunol., 2012, 188, 5 2127–2135

    Article  CAS  PubMed  Google Scholar 

  211. M. Cohen-Lahav, S. Shany, D. Tobvin, C. Chaimovitz and A. Douvdevani, Vitamin D decreases NFkappaB activity by increasing IkappaBalpha levels, Nephrol. Dial. Transplant., 2006, 21, 4 889–897

    Article  CAS  PubMed  Google Scholar 

  212. S. Hansdottir, M. M. Monick, N. Lovan, L. Powers, A. Gerke and G. W. Hunninghake, Vitamin D decreases respiratory syncytial virus induction of NF-kappaB-linked chemokines and cytokines in airway epithelium while maintaining the antiviral state, J. Immunol., 2010, 184, 2 965–974

    Article  CAS  PubMed  Google Scholar 

  213. A. K. Coussens, R. J. Wilkinson and A. R. Martineau, Phenylbutyrate Is Bacteriostatic against Mycobacterium tuberculosis and Regulates the Macrophage Response to Infection, Synergistically with 25-Hydroxy-Vitamin D3, PLoS Pathog., 2015, 11, 7 e1005007

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  214. M. C. Gauzzi, C. Purificato, K. Donato, Y. Jin, L. Wang, K. C. Daniel, et al., Suppressive effect of 1alpha,25-dihydroxyvitamin D3 on type I IFN-mediated monocyte differentiation into dendritic cells: impairment of functional activities and chemotaxis, J. Immunol., 2005, 174, 1 270–276

    Article  CAS  PubMed  Google Scholar 

  215. G. Penna and L. Adorini, 1 Alpha,25-dihydroxyvitamin D3 inhibits differentiation, maturation, activation, and survival of dendritic cells leading to impaired alloreactive T cell activation, J. Immunol., 2000, 164, 5 2405–2411

    Article  CAS  PubMed  Google Scholar 

  216. A. Giulietti, E. van Etten, L. Overbergh, K. Stoffels, R. Bouillon and C. Mathieu, Monocytes from type 2 diabetic patients have a pro-inflammatory profile. 1,25-Dihydroxyvitamin D(3) works as anti-inflammatory, Diabetes Res. Clin. Pract., 2007, 77, 1 47–57

    Article  CAS  PubMed  Google Scholar 

  217. A. K. Coussens, R. J. Wilkinson, Y. Hanifa, V. Nikolayevskyy, P. T. Elkington, K. Islam, et al., Vitamin D accelerates resolution of inflammatory responses during tuberculosis treatment, Proc. Natl. Acad. Sci. U. S. A., 2012, 109, 38 15449–15454

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. G. Nunnari, P. Fagone, F. Lazzara, A. Longo, D. Cambria, G. Di Stefano, et al., Vitamin D3 inhibits TNFalpha-induced latent HIV reactivation in J-LAT cells, Mol. Cell. Biochem., 2016, 418, 1–2 49–57

    Article  CAS  PubMed  Google Scholar 

  219. D. Boraschi, D. Lucchesi, S. Hainzl, M. Leitner, E. Maier, D. Mangelberger, et al., IL-37: a new anti-inflammatory cytokine of the IL-1 family, Eur. Cytokine Netw., 2011, 22, 3 127–147

    Article  CAS  PubMed  Google Scholar 

  220. M. F. Nold, C. A. Nold-Petry, J. A. Zepp, B. E. Palmer, P. Bufler and C. A. Dinarello, IL-37 is a fundamental inhibitor of innate immunity, Nat. Immunol., 2010, 11, 11 1014–1022

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. A. W. Pedersen, K. Holmstrom, S. S. Jensen, D. Fuchs, S. Rasmussen, P. Kvistborg, et al., Phenotypic and functional markers for 1alpha,25-dihydroxyvitamin D(3)-modified regulatory dendritic cells, Clin. Exp. Immunol., 2009, 157, 1 48–59

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. L. Szeles, G. Keresztes, D. Torocsik, Z. Balajthy, L. Krenacs, S. Poliska, et al., 1,25-dihydroxyvitamin D3 is an autonomous regulator of the transcriptional changes leading to a tolerogenic dendritic cell phenotype, J. Immunol., 2009, 182, 4 2074–2083

    Article  CAS  PubMed  Google Scholar 

  223. R. Thien, K. Baier, P. Pietschmann, M. Peterlik and M. Willheim, Interactions of 1 alpha,25-dihydroxyvitamin D3 with IL-12 and IL-4 on cytokine expression of human T lymphocytes, J. Allergy Clin. Immunol., 2005, 116, 3 683–689

    Article  CAS  PubMed  Google Scholar 

  224. J. Smolders, P. Menheere, M. Thewissen, E. Peelen, J. W. Tervaert, R. Hupperts, et al., Regulatory T cell function correlates with serum 25-hydroxyvitamin D, but not with 1,25-dihydroxyvitamin D, parathyroid hormone and calcium levels in patients with relapsing remitting multiple sclerosis, J. Steroid Biochem. Mol. Biol., 2010, 121, 1–2 243–246

    Article  CAS  PubMed  Google Scholar 

  225. A. Coussens, P. M. Timms, B. J. Boucher, T. R. Venton, A. T. Ashcroft, K. H. Skolimowska, et al., 1alpha,25-dihydroxyvitamin D inhibits matrix metalloproteinases induced by Mycobacterium tuberculosis infection, Immunology, 2009, 127, 4 539–548

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. P. E. Van den Steen, I. Van Aelst, S. Starckx, K. Maskos, G. Opdenakker and A. Pagenstecher, Matrix metalloproteinases, tissue inhibitors of MMPs and TACE in experimental cerebral malaria, Lab. Invest., 2006, 86, 9 873–888

    Article  PubMed  CAS  Google Scholar 

  227. P. T. Elkington, J. M. D’Armiento and J. S. Friedland, Tuberculosis immunopathology: the neglected role of extracellular matrix destruction, Sci. Transl. Med., 2011, 3, 71 71ps76

    Article  Google Scholar 

  228. J. S. Mort, G. R. Dodge, P. J. Roughley, J. Liu, S. J. Finch, G. DiPasquale, et al., Direct evidence for active metalloproteinases mediating matrix degradation in interleukin 1-stimulated human articular cartilage, Matrix, 1993, 13, 2 95–102

    Article  CAS  PubMed  Google Scholar 

  229. P. Van Lint and C. Libert, Chemokine and cytokine processing by matrix metalloproteinases and its effect on leukocyte migration and inflammation, J. Leukoc. Biol., 2007, 82, 6 1375–1381

    Article  PubMed  CAS  Google Scholar 

  230. G. W. Hunninghake, J. M. Davidson, S. Rennard, S. Szapiel, J. E. Gadek and R. G. Crystal, Elastin fragments attract macrophage precursors to diseased sites in pulmonary emphysema, Science, 1981, 212, 4497 925–927

    Article  CAS  PubMed  Google Scholar 

  231. G. S. Tjabringa, J. Aarbiou, D. K. Ninaber, J. W. Drijfhout, O. E. Sorensen, N. Borregaard, et al., The antimicrobial peptide LL-37 activates innate immunity at the airway epithelial surface by transactivation of the epidermal growth factor receptor, J. Immunol., 2003, 171, 12 6690–6696

    Article  CAS  PubMed  Google Scholar 

  232. d. Yang, Q. Chen, A. P. Schmidt, G. M. Anderson, J. M. Wang, J. Wooters, et al., LL-37, the neutrophil granule- and epithelial cell-derived cathelicidin, utilizes formyl peptide receptor-like 1 (FPRL1) as a receptor to chemoattract human peripheral blood neutrophils, monocytes, and T cells, J. Exp. Med., 2000, 192, 7 1069–1074

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  233. F. Niyonsaba, K. Iwabuchi, H. Matsuda, H. Ogawa and I. Nagaoka, Epithelial cell-derived human beta-defensin-2 acts as a chemotaxin for mast cells through a pertussis toxin-sensitive and phospholipase C-dependent pathway, Int. Immunol., 2002, 14, 4 421–426

    Article  CAS  PubMed  Google Scholar 

  234. D. Yang, Q. Chen, O. Chertov and J. J. Oppenheim, Human neutrophil defensins selectively chemoattract naive T and immature dendritic cells, J. Leukoc. Biol., 2000, 68, 1 9–14

    CAS  PubMed  Google Scholar 

  235. D. Yang, O. Chertov, S. N. Bykovskaia, Q. Chen, M. J. Buffo, J. Shogan, et al., Beta-defensins: linking innate and adaptive immunity through dendritic and T cell CCR6, Science, 1999, 286, 5439 525–528

    Article  CAS  PubMed  Google Scholar 

  236. N. F. Crum-Cianflone, S. Won, R. Lee, T. Lalani, A. Ganesan, T. Burgess, et al., Vitamin D levels and influenza vaccine immunogenicity among HIV-infected and HIV-uninfected adults, Vaccine, 2016, 34, 41 5040–5046

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  237. C. Cooper, A. Thorne, G. Canadian Hiv Trials Network Ctn Influenza Vaccine Research, Vitamin D supplementation does not increase immunogenicity of seasonal influenza vaccine in HIV-infected adults, HIV Clin. Trials, 2011, 12, 5 275–276

    Article  CAS  PubMed  Google Scholar 

  238. M. E. Sundaram, H. K. Talbot, Y. Zhu, M. R. Griffin, S. Spencer, D. K. Shay, et al., Vitamin D is not associated with serologic response to influenza vaccine in adults over 50 years old, Vaccine, 2013, 31, 16 2057–2061

    Article  CAS  PubMed  Google Scholar 

  239. M. Science, J. L. Maguire, M. L. Russell, M. Smieja, S. D. Walter and M. Loeb, Serum 25-hydroxyvitamin d level and influenza vaccine immunogenicity in children and adolescents, PLoS One, 2014, 9, 1 e83553

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  240. M. K. Lalor, S. Floyd, P. Gorak-Stolinska, R. E. Weir, R. Blitz, K. Branson, et al., BCG vaccination: a role for vitamin D?, PLoS One, 2011, 6, 1 e16709

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  241. J. W. Hsu, P. N. Yin, R. Wood, J. Messing, E. Messing and Y. F. Lee, 1 alpha, 25-dihydroxylvitamin D3 promotes Bacillus Calmette-Guerin immunotherapy of bladder cancer, Oncotarget, 2013, 4, 12 2397–2406

    Article  PubMed  PubMed Central  Google Scholar 

  242. M. Green, Cod liver oil and tuberculosis, Br. Med. J., 2011, 343, d7505

    Article  Google Scholar 

  243. H. M. Mackay, H. M. Linford, M. Mitman and M. H. Wild, The therapeutic value of vitamins A and D in measles, Arch. Dis. Child., 1936, 11, 63 127–142

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  244. A. R. Martineau, P. M. Timms, G. H. Bothamley, Y. Hanifa, K. Islam, A. P. Claxton, et al., High-dose vitamin D(3) during intensive-phase antimicrobial treatment of pulmonary tuberculosis: a double-blind randomised controlled trial, Lancet, 2011, 377, 9761 242–250

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  245. J. Sautet, J. Vuillet and G. Arnaud, [Effects of the immediate adjunction of cod liver oil or vitamin D and calcium biphosphate to antimalarial drugs used in the treatment of Plasmodium berghei infections. II], Bull. Soc. Pathol. Exot. Filiales, 1957, 50, 1 44–49

    CAS  PubMed  Google Scholar 

  246. A. R. Martineau, R. J. Wilkinson, K. A. Wilkinson, S. M. Newton, B. Kampmann, B. M. Hall, et al., A single dose of vitamin D enhances immunity to mycobacteria, Am. J. Respir. Crit. Care Med., 2007, 176, 2 208–213

    Article  CAS  PubMed  Google Scholar 

  247. J. Xia, L. Shi, L. Zhao and F. Xu, Impact of vitamin D supplementation on the outcome of tuberculosis treatment: a systematic review and meta-analysis of randomized controlled trials, Chin. Med. J., 2014, 127, 17 3127–3134

    PubMed  Google Scholar 

  248. N. Tukvadze, E. Sanikidze, M. Kipiani, G. Hebbar, K. A. Easley, N. Shenvi, et al., High-dose vitamin D3 in adults with pulmonary tuberculosis: a double-blind randomized controlled trial, Am. J. Clin. Nutr., 2015, 102, 5 1059–1069

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  249. J. Rathored, S. K. Sharma, B. Singh, J. N. Banavaliker, V. Sreenivas, A. K. Srivastava, et al., Risk and outcome of multidrug-resistant tuberculosis: vitamin D receptor polymorphisms and serum 25(OH)D, Int. J. Tuberc. Lung Dis., 2012, 16, 11 1522–1528

    Article  CAS  PubMed  Google Scholar 

  250. F. R. Sabin, C. A. Doan and R. S. Cunningham, Studies of the Blood in Experimental Tuberculosis: The Monocyte–Lymphocyte Ratio; the Anemia-Leucopenia Phase, Transactions of the 22nd Annual Meeting of the National Tuberculosis Association, 1926, 22, 252–256

    Google Scholar 

  251. A. K. Coussens, A. R. Martineau and R. J. Wilkinson, Anti-Inflammatory and Antimicrobial Actions of Vitamin D in Combating TB/HIV, Scientifica, 2014, 2014, 903680

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  252. A. P. Steenhoff, J. I. Schall, J. Samuel, B. Seme, M. Marape, B. Ratshaa, et al., Vitamin D(3)supplementation in Batswana children and adults with HIV: a pilot double blind randomized controlled trial, PLoS One, 2015, 10, 2 e0117123

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  253. L. Coelho, S. W. Cardoso, P. M. Luz, R. M. Hoffman, L. Mendonca, V. G. Veloso, et al., Vitamin D3 supplementation in HIV infection: effectiveness and associations with antiretroviral therapy, Nutr. J., 2015, 14, 81

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  254. V. A. Stallings, J. I. Schall, M. L. Hediger, B. S. Zemel, F. Tuluc, K. A. Dougherty, et al., High-dose vitamin D3 supplementation in children and young adults with HIV: a randomized, placebo-controlled trial, Pediatr. Infect. Dis. J., 2015, 34, 2 e32–e40

    Article  PubMed  PubMed Central  Google Scholar 

  255. V. Giacomet, A. Vigano, V. Manfredini, C. Cerini, G. Bedogni, S. Mora, et al., Cholecalciferol supplementation in HIV-infected youth with vitamin D insufficiency: effects on vitamin D status and T-cell phenotype: a randomized controlled trial, HIV Clin. Trials, 2013, 14, 2 51–60

    Article  CAS  PubMed  Google Scholar 

  256. A. Nimer and A. Mouch, Vitamin D improves viral response in hepatitis C genotype 2–3 naive patients, World J. Gastroenterol., 2012, 18, 8 800–805

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  257. M. Atsukawa, A. Tsubota, N. Shimada, K. Yoshizawa, H. Abe, T. Asano, et al., Effect of native vitamin D3 supplementation on refractory chronic hepatitis C patients in simeprevir with pegylated interferon/ribavirin, Hepatol. Res., 2016, 46, 5 450–458

    Article  CAS  PubMed  Google Scholar 

  258. E. Falleti, D. Bitetto, C. Fabris, G. Fattovich, A. Cussigh, S. Cmet, et al., Vitamin D binding protein gene polymorphisms and baseline vitamin D levels as predictors of antiviral response in chronic hepatitis C, Hepatology, 2012, 56, 5 1641–1650

    Article  CAS  PubMed  Google Scholar 

  259. E. Rosjo, A. Lossius, N. Abdelmagid, J. C. Lindstrom, M. T. Kampman, L. Jorgensen, et al., Effect of high-dose vitamin D3 supplementation on antibody responses against Epstein-Barr virus in relapsing-remitting multiple sclerosis, Mult. Scler., 2016, 10.1177/1352458516654310

    Google Scholar 

  260. A. Najafipoor, R. Roghanian, S. H. Zarkesh-Esfahani, M. Bouzari and M. Etemadifar, The beneficial effects of vitamin D3 on reducing antibody titers against Epstein-Barr virus in multiple sclerosis patients, Cell. Immunol., 2015, 294, 1 9–12

    Article  CAS  PubMed  Google Scholar 

  261. P. Bergman, A. U. Lindh, L. Bjorkhem-Bergman and J. D. Lindh, Vitamin D and Respiratory Tract Infections: A Systematic Review and Meta-Analysis of Randomized Controlled Trials, PLoS One, 2013, 8, 6 e65835

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  262. D. A. Jolliffe, C. J. Griffiths and A. R. Martineau, Vitamin D in the prevention of acute respiratory infection: systematic review of clinical studies, J. Steroid Biochem. Mol. Biol., 2013, 136, 321–329

    Article  CAS  PubMed  Google Scholar 

  263. A. E. Reeme and R. T. Robinson, Dietary Vitamin D3 Suppresses Pulmonary Immunopathology Associated with Late-Stage Tuberculosis in C3HeB/FeJ Mice, J. Immunol., 2016, 196, 3 1293–1304

    Article  CAS  PubMed  Google Scholar 

  264. B. R. Becklund, K. S. Severson, S. V. Vang and H. F. DeLuca, UV radiation suppresses experimental autoimmune encephalomyelitis independent of vitamin D production, Proc. Natl. Acad. Sci. U. S. A., 2010, 107, 14 6418–6423

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  265. S. Gorman, N. M. Scott, D. H. Tan, C. E. Weeden, R. C. Tuckey, J. L. Bisley, et al., Acute erythemal ultraviolet radiation causes systemic immunosuppression in the absence of increased 25-hydroxyvitamin D3 levels in male mice, PLoS One, 2012, 7, 9 e46006

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  266. M. Ghoreishi, P. Bach, J. Obst, M. Komba, J. C. Fleet and J. P. Dutz, Expansion of antigen-specific regulatory T cells with the topical vitamin d analog calcipotriol, J. Immunol., 2009, 182, 10 6071–6078

    Article  CAS  PubMed  Google Scholar 

  267. C. R. Howson, Heliotherapy in Pulmonary Tuberculosis-Its Possibilities and Dangers, Calif. West. Med., 1928, 29, 1 25–30

    CAS  Google Scholar 

  268. W. B. Grant and E. Giovannucci, The possible roles of solar ultraviolet-B radiation and vitamin D in reducing case-fatality rates from the 1918–1919 influenza pandemic in the United States, Dermatoendocrinology, 2009, 1, 4 215–219

    Article  CAS  Google Scholar 

  269. M. Hartley, S. Hoare, F. E. Lithander, R. E. Neale, P. H. Hart, S. Gorman, et al., Comparing the effects of sun exposure and vitamin D supplementation on vitamin D insufficiency, and immune and cardio-metabolic function: the Sun Exposure and Vitamin D Supplementation (SEDS) Study, BMC Public Health, 2015, 15, 115

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  270. K. Soontrapa, T. Honda, D. Sakata, C. Yao, T. Hirata, S. Hori, et al., Prostaglandin E2-prostaglandin E receptor subtype 4 (EP4) signaling mediates UV irradiation-induced systemic immunosuppression, Proc. Natl. Acad. Sci. U. S. A., 2011, 108, 16 6668–6673

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  271. K. Loser, A. Mehling, S. Loeser, J. Apelt, A. Kuhn, S. Grabbe, et al., Epidermal RANKL controls regulatory T-cell numbers via activation of dendritic cells, Nat. Med., 2006, 12, 12 1372–1379

    Article  CAS  PubMed  Google Scholar 

  272. S. N. Byrne, C. Beaugie, C. O’Sullivan, S. Leighton and G. M. Halliday, The immune-modulating cytokine and endogenous Alarmin interleukin-33 is upregulated in skin exposed to inflammatory UVB radiation, Am. J. Pathol., 2011, 179, 1 211–222

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  273. R. Chacon-Salinas, L. Chen, A. D. Chavez-Blanco, A. Y. Limon-Flores, Y. Ma and S. E. Ullrich, An essential role for platelet-activating factor in activating mast cell migration following ultraviolet irradiation, J. Leukoc. Biol., 2014, 95, 1 139–148

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  274. S. N. Byrne, A. Y. Limon-Flores and S. E. Ullrich, Mast cell migration from the skin to the draining lymph nodes upon ultraviolet irradiation represents a key step in the induction of immune suppression, J. Immunol., 2008, 180, 7 4648–4655

    Article  CAS  PubMed  Google Scholar 

  275. R. Chacon-Salinas, A. Y. Limon-Flores, A. D. Chavez-Blanco, A. Gonzalez-Estrada and S. E. Ullrich, Mast cell-derived IL-10 suppresses germinal center formation by affecting T follicular helper cell function, J. Immunol., 2011, 186, 1 25–31

    Article  CAS  PubMed  Google Scholar 

  276. S. N. Byrne and G. M. Halliday, B cells activated in lymph nodes in response to ultraviolet irradiation or by interleukin-10 inhibit dendritic cell induction of immunity, J. Invest. Dermatol., 2005, 124, 3 570–578

    Article  CAS  PubMed  Google Scholar 

  277. T. Schwarz and A. Schwarz, Molecular mechanisms of ultraviolet radiation-induced immunosuppression, Eur. J. Cell Biol., 2011, 90, 6–7 560–564

    Article  CAS  PubMed  Google Scholar 

  278. A. Maeda, S. Beissert, T. Schwarz and A. Schwarz, Phenotypic and functional characterization of ultraviolet radiation-induced regulatory T cells, J. Immunol., 2008, 180, 5 3065–3071

    Article  CAS  PubMed  Google Scholar 

  279. A. M. Moodycliffe, D. Nghiem, G. Clydesdale and S. E. Ullrich, Immune suppression and skin cancer development: regulation by NKT cells, Nat. Immunol., 2000, 1, 6 521–525

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Anna K. Coussens.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Abhimanyu, Coussens, A.K. The role of UV radiation and vitamin D in the seasonality and outcomes of infectious disease. Photochem Photobiol Sci 16, 314–338 (2017). https://doi.org/10.1039/c6pp00355a

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1039/c6pp00355a

Navigation