Está en la página 1de 22

Immunology and Cell Biology (2001) 79, 547–568

Review Article
Ultraviolet light induced injury: Immunological and inflammatory
effects
G AV I N J C LY D E S DA L E , * G E O F F R E Y W DA N D I E † a n d H KO N R A D M U L L E R

Discipline of Pathology, University of Tasmania, Hobart, Tasmania, Australia

Summary This article reviews many of the complex events that occur after cutaneous ultraviolet (UV) exposure.
The inflammatory changes of acute exposure of the skin include erythema (sunburn), the production of inflam-
matory mediators, alteration of vascular responses and an inflammatory cell infiltrate. Damage to proteins and
DNA accumulates within skin cells and characteristic morphological changes occur in keratinocytes and other skin
cells. When a cell becomes damaged irreparably by UV exposure, cell death follows via apoptotic mechanisms.
Alterations in cutaneous and systemic immunity occur as a result of the UV-induced inflammation and damage,
including changes in the production of cytokines by keratinocytes and other skin-associated cells, alteration of
adhesion molecule expression and the loss of APC function within the skin. These changes lead to the generation
of suppressor T cells, the induction of antigen-specific immunosuppression and a lowering of cell-mediated immu-
nity. These events impair the immune system’s capacity to reject highly antigenic skin cancers. This review gives
an overview of the acute inflammatory and immunological events associated with cutaneous UV exposure, which
are important to consider before dealing with the complex interactions that occur with chronic UV exposure,
leading to photocarcinogenesis.

Key words: inflammation, immunosuppression, skin, ultraviolet rays.

Introduction The peak of UV-induced carcinogenicity has been shown


to lie within the UVB portion of the UV spectrum (reviewed
Ultraviolet (UV) radiation forms part of the electromagnetic
by de Gruijl2). As the primary consequence of ozone deple-
spectrum with wavelengths between 200 nm and 400 nm and
tion is an increased amount of UVB, and potentially some
is divided into three further components, UVA (320–400 nm),
UVC, reaching the earth’s surface, it may, in turn, lead to an
UVB (280–320 nm) and UVC (200–280 nm).1 Solar UVC is
increased incidence of skin cancer and eye damage.3 Com-
absorbed by ozone in the stratosphere and its relevance to
bined with the possibility that UVB alters immune responses
biological systems is limited, thus, the division between UVC
to infectious agents (reviewed by Chapman4), this has
and UVB was established at 280 nm. Discussion of UVC
brought the detrimental effects of UV exposure to the fore-
effects on the skin are, therefore, kept to a minimum is this
front of scientific discussion. This is especially relevant in
review. The energy carried by each portion of the spectrum is
Australia, where the highest incidence of any single cancer,
inversely related to its wavelength so UVC > UVB > UVA.
basal cell carcinoma, occurs.5 At times Australia also lies
However, the depth of penetration of UV light into the skin
within or near the ‘hole in the ozone layer’, perhaps, further
increases with increasing wavelength.
increasing the risk of developing such cancers in this
The skin is the organ most susceptible to damage by UV
country.6
light as it is directly exposed. Ultraviolet exposure of the skin
has a number of biological effects, many of which are detri-
mental. The following sections deal with the effects of UV Ultraviolet light methodology
light on the skin, concentrating on the inflammatory and
A variety of light sources have been used to study the
immunological events in a range of animal, human and in
changes that occur after UV exposure of the skin. These vary
vitro models.
considerably in their emission spectra and have advantages
and disadvantages relative to each experimental model exam-
ined. A summary of different UV sources used in cutaneous
Correspondence: Prof. HK Muller, Discipline of Pathology, research is given in Table 1.
University of Tasmania, PO Box 252-29, Hobart, Tas. 7001, Aust- Usually UV treatment is given as a dose per unit area.
ralia. Email: Konrad.Muller@utas.edu.au
* However, this is often referred to in terms of minimal ery-
Present address: Dr GJ Clydesdale, Pest Animal CRC, CSE Sus-
tainable Ecosystems, PO Box 284, Canberra, ACT 2601, Australia. themal dose (MED), with little or no reference to the actual

Present address: Dr GW Dandie, Child Health Research Institute dose received. Stern and Urbach8 stated that ‘it is not always
Inc., 7th Floor, Clarence Rieger Building, Women’s and Children’s clear whether “minimal” refers to the shade of erythema pro-
Hospital, 72 King William Road, North Adelaide, SA 5006, Australia. duced or to the least amount in energy in one of a series of
Received 8 August 2001; accepted 8 August 2001. increasing doses that produces erythema’. Stern and Urbach8
548 GJ Clydesdale et al.

Table 1 Ultraviolet light sources used for research (adapted from Anderson7)

Light source UV emission Erythemal dose Advantages Disadvantages Peak output


spectra (nm) (time) (nm)

Terrestrial 290–400 20 min at Economical, Variable output, 400


sunlight latitude 40° large field hard to predict
June midday
Carbon arc 200–400 5–30 s at 25 cm Emission spectra Variable output, 400
close to sunlight large and bulky
Cold quartz Discontinuous, 30 s at 25 cm No increase Not relevant to 253.7
253.7 in pigment, sunlight as totally UVC
predominantly inexpensive
Hot quartz Discontinuous 30–60 s at 45 cm Inexpensive, Warm up, discontinuous 365
254,265,297 303 relatively intense UVB emission (not natural)
313 and 365
Fluorescent sun 270–390 60–120 s at 25 cm Large field size Lamp output 313
lamps when used in banks, decreases with time
intense UVB, inexpensive (70% after 500 h).
Not natural (some UVC)
Fluorescent 310–400 15–16 min at 25 cm Stimulation of pigment Long exposure time 365
black light production to generate erythema
Narrow band 311–312 5–9 min at 25 cm Exposure is less Not natural, long 312
(UVB) hazardous, study of exposure time to
specific wavelengths generate erythema
Xenon arc 290–400 12 s at 55 cm Emission spectra close Expensive, technical, 400
to natural sunlight bulky, small field size

Table 2 Minimal erythemal dose for a variety of ultraviolet sources and species

Species Average dose Main UV Source of UV irradiation Reference


for MED (J/m2) range

Guinea pigs 900 UVB Monochromatic 295 nm Eaglstein et al.12


Guinea pigs 260 UVC Monochromatic 250 nm Eaglstein et al.12
Guinea pigs 246 UVB Sunlamp Morison et al.13
Human dermal 30 (10% UVB UVB Sunlamp Cornelius et al.14
microvascular reaches the dermis)
endothelial cells
Humans 5 × 105 UVA Xenon Arc (= solar simulator) Gilchrest et al.15
Humans 8.6 × 105 UVA Xenon arc (345 filter) Kumakiri et al.16
Humans 1.2 × 106 UVA UVA sunlamp (330 nm & UG1 filter) Baadsgaard et al.17
Humans 200 UVB Monochromatic 300 nm Everett et al.18
Humans 200–300 UVB Monochromatic 300 nm Norris et al.19
Humans 264 UVB Xenon arc (345 filter) Kaidbey et al.20
Humans 291–327 UVB Sunlamp Cooper et al.21
Humans 300 UVB Sunlamp Cornelius et al.14
Humans 450 UVB Hot quartz Gilchrest et al.22
Humans 975 UVB T12 (307 nm filter) Baadsgaard et al.17
C3H Mice 1600 UVB TL20 El-Ghorr et al.23

subsequently commented that variables such as time to gen- produced and stored. Table 2 gives a short summary illus-
erate MED or the spectral output of the light source used trating the varying doses required to generate MED in differ-
were rarely mentioned. Recent publications have begun to ent species and strains and the sources of UV radiation.
correct these oversights, thus allowing easier comparison Generally, most researchers currently use sunlamps in
between different investigations. In humans the dose/expo- studying the effects of UV light on the immune system. The
sure time to reach MED varies from individual to individual use of sunlamps is widespread due to their relatively low cost
due to genetic differences reflected in the amount of melanin and high UVB output. It has been suggested that the UVC
Ultraviolet light induced injury 549

output (non-solar UV) by FS40 sunlamps is sufficient to consideration as the time course of UVA erythema in humans
affect the mixed lymphocyte reaction (MLR) and epidermal differs from UVB, being maximal at 72 h compared to
cell–lymphocyte reaction (ELR) experiments.9 A study by 24–48 h for UVB.38 Adaptive changes are also observed in
Roberts et al.10 also found that energy sources with spectral the skin after exposure to UV light. These include increased
emissions similar to sunlight are required when evaluating vascularization,39 skin thickening and melanogenesis
the effects of sunscreens on UV-induced immune suppres- (tanning).40 Increased skin thickening is due both to epider-
sion. Conversely, Cooper et al.11 have shown that the mal hyperplasia and increased thickness of the stratum
immunosuppression arising from 4 MED of both FS40 sun- corneum.41 Increased proliferation in keratinocytes, assessed
lamps and solar simulator lamps is identical, thus validating by the presence of proliferating cell nuclear antigen, is
the extensive use of sunlamps in this area of research. maximal 48 h after UV exposure of the skin and parallels
However, there is a growing trend towards using solar simu- hyperplasia.42 The hyperplastic effects of a single exposure of
lated light, as this appears more relevant to studies on mouse skin can last for up to 2 weeks.42 These changes result
immune effects, sunscreen use and carcinogenesis that can be in an overall reduction in the barrier function of the skin.43,44
related to actual sunlight exposure. Increased melanin production provides increased protection
against the harmful effects of UV exposure,45 to the extent
that black people with a high melanin content, have a MED
Inflammatory response 33-fold higher than their Caucasian counterparts.46
Many factors have been suggested as potential mediators
Vascular events
for the erythemal response of skin to UV light, yet most of
The initial inflammatory response of the skin to UV irradia- these have been found to have no or only a minor effect.
tion is vasodilation of cutaneous blood vessels resulting in Logan and Wilhelm31 thought that the early erythemal
erythema. Finsen24 in 1899 postulated that erythema was response to UV light was similar to that of thermal injury
caused by UV exerting a direct effect on the vessels. An alter- mediated by histamine. But, the addition of antihistamines
nate theory by Partington,25 was that this effect was produced did not affect the induction of erythema in either the guinea
by soluble vasoactive mediators acting on the vessels, pig or rabbit models examined. The early response observed
although no direct evidence was provided for either hypoth- in rats was mediated by serotonin (5-HT), as the addition of
esis. The time course for the production of erythema is bi- an antagonist (BOL-148) blocked the induction of erythema,
phasic with an initial immediate erythema observed before while the addition of antihistamines had no effect. However
the completion of exposure.26 This is followed by a further H1 and H2 receptor antagonists were able to eliminate the
increase, usually of greater magnitude, commencing 1 h later, early erythemal response in guinea pigs, but again not
with a peak at 24–48 h. The reddening that results at this the delayed erythemal response.47 Greaves and Sondergaard48
stage is also due to an increase in vasodilation.26 Later studies have shown that UVB erythema in humans is linked with the
using more sensitive instruments, which measured reflect- action of a smooth muscle contracting agent. This agent was
ance, demonstrated that vasodilation preceded visually per- clearly different from histamine, bradykinin, serotonin or
ceptible erythema.27 The degree of erythema observed after acetylcholine and was shown to be a fatty acid, possibly a
treatment with UV light increases proportionally with dose prostaglandin (PG). Indeed, an increase in the production of
until at least 15 MED,28 depending on skin type. Eventually, PGE2 and PGF2α and their precursor arachidonate was
at higher doses, blistering and other signs of gross damage evident 24 h after UVB exposure of the skin.49 Inhibitors of
are observed. Erythemal responses are also influenced by PG synthesis, including aspirin50 and indomethacin,51 have
environmental factors, including heat, wind,29 humidity and been previously noted to decrease the erythemal response but
water immersion.30 not eliminate it, providing indirect evidence for the involve-
Logan and Wilhelm26,31–33 investigated the erythemal reac- ment of PG. Woodward and Owen52 found that increases in
tion in a variety of animal models including rats, rabbits and skin temperature, vascular permeability and water content of
guinea pigs. They looked at various aspects, including the skin were substantially reduced, but not eliminated, by
vascular permeability, tissue leukocytosis and the production indomethacin, strongly implicating PG as a mediator of these
of erythema and found variation in the exposure times physiological effects of cutaneous UV exposure. The appli-
required to provoke erythema in the different species. Vascu- cation of indomethacin topically, intradermally or orally, has
lar permeability was found to follow the same bi-phasic been shown not to affect UVA or 8-methoxypsoralen/UVA
pattern as the production of erythema, with an initial increase treatment (PUVA) induced erythema.53 Thus, UVA-induced
followed by a return to normal levels after 30 min and then erythema is not due to a PG-mediated event.
followed, in turn, by a further substantial increase in vascular Recently, nitric oxide (NO) was shown to be produced by
permeability. This general pattern held for the three species keratinocytes after UVB irradiation.54 Such NO release is
examined, although the timing varied slightly.31 This pattern dose-dependent and keratinocytes constitutively express the
has also been observed in humans.34 However, while Cotran enzyme required to synthesize NO. The same study demon-
and Pathak35 confirmed this in rats and guinea pigs, the same strated that in the guinea pig an inhibitor of NO synthetase
pattern was not evident in hairless mice, which showed only (L-NMMA) resulted in a sun (UV) protection factor (SPF) of
the characteristic delayed increase in vascular permeability. 8.71. The authors concluded that this might be a major part
Blood flow changes appear to be similar for the UV- of the integrated inflammatory response to UV light leading
induced erythemal response using both germicidal lamps to vasodilation and erythema. Current evidence suggests that
(UVC) and sunlamps (UVB/UVA).36,37 However, when DNA is the chromophore for erythema, as the action spec-
studying UVA-induced erythema, timing is an important trum for the frequency of pyrimidine dimers in vivo closely
550 GJ Clydesdale et al.

matches the action spectrum for erythema up to 334 nm.55 Exposure in vitro of an epidermal-derived cell line to both
This conclusion has been strongly supported by work con- UVA and UVB produced cytoskeletal blebbing, this being a
ducted in the South American Opossum, Monodelphis characteristic of oxidative damage,69 and perhaps reflects the
domestica, which has shown photoreactivation of DNA via major mechanism for UVA-induced damage.
the actions of photolyase, a DNA repair enzyme, which leads As sunlight contains both UVA and UVB it is important
to the reduction of erythema.56 This in turn further implicates that the cytotoxic effects of the UVA portion of the spectrum
pyrimidine dimers as the major DNA lesion involved with the are not ignored, especially as a study by Tyrrell and Pidoux70
production of erythema. Further support for this proposal suggests that UVA-induced cytotoxicity may be just as, if not
comes from studies that involved another DNA repair more, important than UVB. When the sun is at its zenith
enzyme specific for dimers, T4 endonuclease V (T4N5). The (10 AM–2 PM) and UVB maximal, UVA still makes up to
application of this enzyme to the skin in liposomes has been 50–60% of the phototoxic wavelengths found, and this is
shown to reduce the production of oedema in hairless mice.57 increased at other times.70 The wavelength of UV light influ-
Thus, the vasodilation and subsequent erythema produced ences the cytotoxicity seen, with UVB > UVA. Generally
after exposure of the skin to ultraviolet light, is not due to the UVB acts mainly in the epidermis, while UVA injury tends to
production of any one substance, but is multifactorial with cause necrosis of the endothelial cells, thus damaging the
NO and PG having key roles. Other inflammatory substances, dermal blood vessels.40,71 However, it should be noted that
including inflammatory cytokines, may have minor, but still 10–15% of UVB penetrates to the dermis and, hence, it is
important, roles in the complicated reaction to UV light. possible for UVB to have a direct effect on microvascular
These, in turn, affect the immune response that may be gen- endothelial cells.72 In vitro studies by Cornelius et al. also
erated from the affected area by altering adhesion molecule support a direct effect of UVB on endothelial cells and direct
and cytokine expression of resident cell populations and the cytotoxic effects on cells in the skin are evident after UVA
induction of non-resident cell types to migrate into the area, exposure.14 Sunburn cells are visible in the epidermis after
resulting in the generation of an ‘inflammatory’-linked UVA irradiation, although fewer in number than the equiva-
immune response. lent dose of UVB.16,73 In contrast, a study using erythemal
doses of UVA only, showed that while sunburn cells were
absent from the epidermis, perinuclear vacuoles were present
Morphology and ultrastructural changes
in LC.15 The same study demonstrated the recruitment of
One consequence of UV exposure of the skin is damage to neutrophils into the UVA-irradiated skin. UVB doses that
epidermal cells.58 This becomes evident as early as 2 h after cause similar amounts of erythema did not appear to induce
UV exposure.31 The earliest indicator of damage is a decrease this change,22 although this has been reported for rodent-
in keratinosomes, which results in the formation of dyskera- based experiments.12 Detailed studies by Hawk et al.74 were
totic cells;59 intracellular oedema can be seen 16–18 h after also in direct contrast to those of Gilchrest’s study;22 Hawk
exposure, followed at 30–48 h by intercellular oedema, et al.74 suggested that the lack of similar findings by
which develops around damaged keratinocytes. Sunburn cells Gilchrest et al.22 was due to ‘failure to obtain biopsy mater-
(damaged keratinocytes) appear shortly before the presence ial at times of significant neutrophil infiltration’. However, it
of oedema is observed.31 must be noted that the Hawk et al.74 study utilized a sunlamp
The sunburn cell was proposed as one of the earliest that had an output, of which approximately one third was
examples of apoptosis.60 Ultraviolet-induced apoptotic cells UVA.
are rapidly phagocytised by the surrounding keratinocytes.61 The peak infiltration of leucocytes after UVB irradiation
Macrophages are also known to bind to and phagocytose occurs at 4–6 h and the response concludes after 48 h.26
apoptotic cells62 and their numbers in the skin increase Studies conducted using guinea pigs showed that the peak in
dramatically after UVB treatment.63 All wavelengths of the dermal leukocytosis occurred before maximum erythema and
UV spectrum, including UVC, are able to induce apoptosis vascular permeability, suggesting a cause and effect relation-
rather than necrosis in Ly-5 murine lymphoma cells.64 There ship. Later studies by the same authors in the rat and rabbit31
appear to be two types of UV-induced apoptosis, an immedi- indicated that lymphocyte infiltration was not as important in
ate type and a more classical delayed apoptosis.65 The latter these animal models and peaked after maximum erythema
is thought to be caused by UV-induced damage of DNA and vascular permeability. Eaglstein et al.,12 suggested that
rather than free radical damage.65 In addition, immediate leucocytes have a role in UVB induced-inflammation in
apoptosis appears to be due to constitutive factors that do not guinea pigs, but not in cases of inflammation caused by UVA
require protein synthesis, while the delayed type (inducible) or UVC. The presence of neutrophils is seen in dermal
apoptosis does require protein synthesis.65 inflammation due to UVB irradiation in humans. These cells
Similar apoptotic changes are noted in epidermal Langer- were observed, a short time after irradiation, gradually
hans cells (LC), which are especially sensitive to the effects increasing in number until at least 48 h post-treatment.74
of UV light,66,67 with damage observed at lower doses than Neutrophils have the potential to enhance the physical
required for keratinocytes. Langerhans cells are more sensi- damage caused by UV light, as they are capable of releasing
tive to UV-induced damage in comparison to keratinocytes a variety of substances, including reactive oxygen intermedi-
for both UVA and UVB wavelengths.66 The degenerative ates, which are damaging to cells and tissues.75
changes for LC and keratinocytes include mitochondrial The mast cell is a cell resident within the dermis, which
swelling and rupture, condensation of the cytoplasm and the has an important role in UV-induced inflammation. Physical
appearance of pyknotic nuclei.58,66–68 The observed cellular changes in mast cells have been noted after UVB exposure,
damage is maximal at 48–72 h for the total UV range.40 including the presence of hypogranulated and degranulated
Ultraviolet light induced injury 551

cells and extracellular granules in the surrounding tissue.76 In death), allowing the p53 mutant to dominate. In association
the lower dermis, mast cell numbers increase after chronic with the hyperplastic status of the skin this leads to tumour
UVB exposure of hairless mice; the biological effect of this promotion.93 There are findings to support this hypothesis,
is unknown.77 The role of mast cells in immunological events with 87.5% of skin tumours in hairless mice subject to
after UV exposure is discussed in later sections. chronic UV exposure being positive for p53 mutations; 52%
of these mutations occurred at codon 270, a hot spot associ-
ated with UV mutations.94
Ultraviolet damage of DNA
DNA damage (pyrimidine dimer formation) has also been
The heterocyclic bases of DNA are a major UV-absorbing proposed as a chromophore for the initiation of systemic
chromophore in the skin.78 The absorption (which is maximal immunosuppression in mice after UV irradiation.95 Photo-
between 260 nm and 265 nm) leads to damage of the DNA, repair of DNA damage is seen in the marsupial Mondelphis
which may result in the generation of tumours. This part of domestica. This animal contains photorepair enzymes that
the review concentrates on the direct physical damage of are activated by visible light and prevent the induction of UV-
DNA by UV light and will touch briefly on some of the induced immunosuppression.96 Treatment of mouse skin with
molecular events taking place as this is currently a rapidly liposomes containing the excision repair enzyme T4 endo-
expanding area of research. nuclease V (T4N5), increases the rate of pyrimidine dimer
DNA strongly absorbs energy in the UVB region of the repair,97 thus, preventing the UV-induced immunosuppression
spectrum and may result in DNA lesions. The main lesions of delayed-type hypersensitivity (DTH) and contact hyper-
induced by UVB exposure are pyrimidine dimers and sensitivity (CHS).95 Kripke et al.95 proposed that DNA
pyrimidine–pyrimidone photoproducts, which make up damage leads to the release of immunoregulatory cytokines.
65%79 and 35%80 of the UVB-induced DNA lesions, respec- Indeed preliminary unpublished data by this group found
tively. Of the pyrimidine dimers formed, 50% are thymine– that downregulation of IL-10 production by a UV-damaged
thymine dimers.81 These lesions have been shown to occur keratinocyte cell line (PAM212) occurred after treatment
in the epidermal basal and suprabasal layers and can be with T4N5 liposomes, thereby, indicating a role for DNA
found in LC.82 Pyrimidine dimer formation is proposed as damage in UV-induced systemic suppression (reviewed by
an initiation step in mutagenesis and tumour formation.83 Vink et al.,98). Local immunosuppression is also manipulated
Mutations in the ras oncogene were mostly found opposite by T4N5 treatment, which prevents LC loss from the epider-
dipyrimidine sequences,84 providing more evidence for the mis and partially reduces local immunosuppression.99
role of pyrimidine dimers in tumour genesis. The exposure Normally, cells with UV-damaged DNA can be observed in
of skin to UV radiation also increases the proto-oncogene the draining lymph node after UV treatment of the skin.
c-fos.85 UVB can induce nuclear factor κB (NFκB), although Treatment of these UV-damaged cells with the photolyase
without the need for chromosomal DNA damage.86 The photorepair enzyme restores their ability to induce a normal
induction of NFκB after UV exposure of the skin, in turn, immune response. This treatment also prevents the induction
leads to the production of the pro-inflammatory cytokines of suppressor cells (reviewed by Vink et al.98). A recent study
IL-1, IL-6 and TNF-α.87 has shown that thymidine dinucleotides alone are sufficient
Ultraviolet light induces mutations in p53, an anti- to induce immunosuppression of CHS and the activation of
oncogene.88 This has been demonstrated in squamous cell the TNF-α gene.100 This further increases support for the
carcinoma,88 however, mutations in p53 seem to be absent hypothesis that DNA damage is the trigger for many events
from melanomas.89 p53 stops progression of cells through the surrounding UV-induced damage of the skin, including
normal cell cycle after UV exposure, thus allowing time for immunosuppression.
the cell to repair DNA damage. As p53 can lead to the induc- Protective measures are employed to defend the body
tion of apoptosis, it is not surprising that in p53 knockout against the toxic effects of cutaneous UV exposure. Two
mice (p53–/–), the induction of sunburn cells is much lower DNA repair pathways exist in humans to eliminate UV-
than in wild-type mice (p53+/+).90 The heterozygote (p53+/–) in induced DNA damage; the global genome repair pathway and
this study displayed an intermediate phenotype, suggesting the transcription-coupled repair pathway. In humans, defi-
that the loss of one functional copy of the p53 gene in a cell ciencies in these pathways leads to increased susceptibility to
is enough to have an effect. In another study, tumour devel- UV-induced skin cancer, as seen in xeroderma pigmentosum
opment occurred after 24 weeks of chronic UV exposure patients deficient in the global genome repair pathway, but
compared to 46 weeks for the wild-type litter mates.91 Thus, not Cockayne syndrome patients who are deficient in the
the loss of a single functional copy of this gene resulted in transcription-coupled repair pathway. The overwhelming
increased susceptibility to UV photocarcinogenesis. More of repair mechanisms by exposure to excessive amounts of
recent evidence confirms that UV-induced apoptosis of DNA damage from UV light can lead to damaged cells being
keratinocyes occurs via the p53/p21/bax pathway, with retained in the epidermis. From recent studies using murine
sequential increases in these genes in the epidermis being models with defects in either transcription-coupled or global
maximal at 12, 24 and 24–72 h, respectively.42 In the same genome repair pathways, it appears that oedema and erythema
study an inverse relationship between bax and bcl-2 expres- are mediated by the transcription-coupled repair pathway.101
sion was noted. The over-expression of bcl-2 in the epidermis Both pathways must be abrogated before increased sensitivity
has been shown to protect cells against UV-induced apopto- to UV immune suppression is observed.102
sis.92 Therefore, sunlight may, by mutating a p53 gene, cause Exposure of human skin to 2 MED of UVB induces
tumour initiation (DNA mutation) and by further exposure, keratinocyte and fibroblast induction of the protective metal-
may delete the normal cells (which undergo programmed cell lothionein genes in vivo.103 The metallothionein proteins are
552 GJ Clydesdale et al.

Table 3 Reduced resistance to infection by cutaneous ultraviolet exposure

Microorganism Effect Model Reference:

Mycobacterium bovis Decreased DTH response Murine Jeevan et al.127


Mycobacterium leprae Decreased DTH response Murine Cestari et al.128
Listeria monocytogenes Decreased DTH response Murine Goettsch et al.129
Candida albicans Decreased DTH response Murine Denkins et al.130
Murine leukemia virus Increased liver damage Murine Brozek et al.131
Rat cytomegalovirus Increased viral load, inflammation and tissue necrosis Rat Goettsch et al.132
Garssen et al.133
Herpes simplex virus-1 Decreased DTH response, increased recrudesence and lesion severity Murine Ross et al.134 Howie et al.135
after secondary infection Ross et al.136
El-Ghorr et al.137
Herpes simplex virus-2 Severity of infection increased plus suppression of DTH and Murine Yasumoto et al.138
lymphocyte proliferation
Herpes labilis Increased lesions and virus shedding Human Rooney et al.139
Borreilia burgdorferi Switch from Th1 to Th2 antibody isotypes and a reduced DTH Murine Brown et al.140
response
Trichinella spiralis Increased parasite load and reduced T-cell immunity to parasite Rat Goettsch et al.132
Goettsch et al.141
Goettsch et al.142

DTH, delayed-type hypersensitivity.

proposed to have a possible protective role in the skin against DTH.114 However, little effect was shown on levels of anti-
photo-oxidative stress,104 which induces strand breaks in body production,115 while some reduction of antibody level
DNA and interferes with the transcription of other genes such was observed by Spellman.116
as c-fos.105 Thus, expression of high levels of metallo- The UV dose used for generating immune suppression
thioneins by basal keratinocytes may protect cells from varies greatly from model to model and antigen to antigen. To
UV-induced DNA damage.103 investigate local immune suppressive effects on APC, a
Hence, UV light is capable of directly damaging DNA single low dose (< 1 kJ/m2) is generally used. To study the
causing mutations that may give rise to tumours, especially effects of systemic immune suppression by UV light, a single
when mutations in oncogene and anti-oncogenes occur. DNA high acute dose can be used or alternatively four exposures to
damage is the primary change required for the initiation of 400 J/m2. However, it also needs to be stated that low-dose
tumours, however, for a tumour to become established a UV can cause systemic immune suppression as well as local,
tumour promotion step is required. This secondary change but the systemic effects develop more slowly.117–119 The dose
may be provided by alteration of immune function and required to suppress DTH or CHS is different for specific
immunosurveillance within the skin after subsequent expo- antigens, so these differing methodologies must be taken into
sure to UV light.106 account when making comparisons between different studies.
Many investigations have been undertaken since the early
discovery of UV-induced suppressor cell activity, aimed at
Ultraviolet-induced immunosuppression
identification and characterization of these cells. Thy1.2+
In 1963 Hanisko and Suskind,107 observed that the contact cells have been shown to abrogate suppressive activity after
hypersensitivity response in skin sensitized to dinitro- UVB exposure. This was found to be specific for the hapten
chlorobenzene (DNCB) was reduced if skin was previously applied through the irradiated skin.120 Treatment with Lyt1
exposed to suberythemal doses of UVB. This important antibodies was shown to remove suppression and allow rejec-
observation was the first of many examining the effects of tion of tumours, suggesting that these cells are required for
UV on the immune system. The next set of important obser- immune suppression.121 The presence of suppressor cells in
vations were made by Kripke,108 who investigated the anti- strains of UV-resistant and UV-sensitive mice indicated that
genicity of UV-induced tumours and showed that the induced mice of the UV-sensitive phenotype were sensitive due to
tumours were highly immunogenic and were only trans- interruption of effector mechanisms, not the induction of
plantable to immunosuppressed hosts. Kripke and Fisher109 suppressor cells.122 This hypothesis was supported by the
found that UV-induced tumours were able to grow in the fact that UVB exposure converts LC from immunologically
primary host due to a systemic alteration in the host’s potent APC, to tolerogenic APC; stimulation of Th1 cells is
immune response to the tumour. This was due to the UV reduced, while the ability to stimulate a Th2 response
exposure received, as UV-irradiated recipients were unable remains.123 Clonal anergy also results in Th1 cells in response
to reject the tumours. This effect was transferable with T to UV-exposed LC.123 This provides a mechanism whereby a
lymphocytes.110 UVB exposure has been shown to result in ‘suppressor’ cell is induced (Th2 cell) and the effector cell
suppression of many immune parameters including lympho- (Th1) is eliminated or anergized. Simon124 demonstrated that
cyte function111 and recirculation,112 CHS responses113 and Th1 cells in draining lymph node cells from normal and
Ultraviolet light induced injury 553

hapten-treated murine skin produced IL-2 and IFN-γ. various light sources and combinations of filters, studies may
However, after UVB exposure and hapten treatment of the now be manipulated to examine the effects of UVA, or indeed
skin these cytokines could not be detected. This implies that specific regions in the UVA range, on the immune system.
the UV-irradiated skin immune system would lose immuno- Although older studies had indicated that UVA exposure
surveillance against tumours or infectious agents requiring a appears to be less immunosuppressive than UVB,147 UVA
cellular response.125 This alteration of the immune response wavelengths greater than 315 nm are weakly immuno-
may be due to the action of IL-10 post-UV exposure.126 suppressive.148 The contrasting effects of UVA and UVB have
Ultraviolet-induced immunosuppression results in many recently been confirmed by a study in which human skin was
aberrant responses to microorganisms, especially those of exposed to equivalent erythemal doses. UVB was shown to
intracellular bacteria and viruses in which immunity relies depress sensitization to the contact allergen diphenylcyclo-
extensively on the cellular arm of the immune system. From propenone, while UVA did not result in decreased immu-
the studies summarized in Table 3, it is evident that decreased nization rates.149 Studies by Reeve et al.150 have shown that
cellular immunity observed after UVB exposure of the skin wavelengths in the UVA region may be photoprotective if
can lead to alterations in the body’s ability to respond to a given before UVB exposure. More recently this group has
variety of viral and intracellular microbes. linked this photoprotective effect to the production of IFN-γ,
Studies on Borrelia burgdorferi infection illustrate the as UVA photoprotection was not seen in IFN-γ knockout
switch from cellular to humoral immunity that is now thought mice.151 Furthermore, this photoprotection was mediated by
to occur after UV exposure.140 Brown et al.140 demonstrated increased levels of the enzyme, heme oxygenase.152 Skov
that following UV exposure and immunization with B. et al.153 found that UVA1 and UVB differentially regulate the
burgdorferi, IgG2a and IgG2b isotypes were downregulated production of TNF-α, with UVA1 causing a downregulation
(Th1 isotypes), while IgG1 was unaffected (Th2 isotype). in the levels of TNF-α in comparison to normal unirradiated
Although the identity of the suppressor cells remains to be skin. This contrasts with UVB-irradiated skin in which an
elaborated, Yagi143 was able to generate a line of Th2-like upregulation of TNF-α is observed. Longuet-Perret et al.154
cells that expressed CD3+CD4+CD8–Vβ7+ from UVB- confirmed these findings in vitro, and have also shown that
pre-irradiated, tetracholorosalicylanilide photosensitized UVA exposure leads to the inhibition of vascular endothelial
mice that had received repeated antigenic stimulation. These cell growth factor (VEGF) production by keratinocytes. The
cells were shown to suppress contact photosensitivity, both in findings of Skov et al.,153 Reeve et al.150 and Longuet-Perret
vivo and in vitro. This activity was neutralized by anti-IL-10 et al.154 suggest that UVA, especially UVA1, exposure of the
mAb, but not by anti-IL-4 mAb. This cell type may reflect the skin may have beneficial effects. In contrast, Damian et al.155
suppressor cell population(s) that occurs in vivo. Recently, have shown that UVA alone is sufficient to downregulate
evidence from a murine model of DTH suggests that a rela- the recall response to nickel in humans, suggesting that the
tively minor T-cell population expressing the NK cell marker, photoprotective effects may be overcome. These findings are
DX5, has been shown to be the cell type responsible for the causing researchers in photobiology to shift away from the
generation of UV-induced immune suppression.144 After UV use of non-solar simulated light sources, such as the fluores-
exposure, one million cells of this phenotype were shown cent sunlamp, the output of which contains low amounts of
to be sufficient to transfer immune suppression to non- UVA relative to UVB in comparison with sunlight.
irradiated mice in an antigen-specific manner. This effect
could be linked to the ability of these cells to produce high
Genetic basis for immunosuppression
levels of IL-4 during the early stages of an immune response.
Ultraviolet-induced immunosuppression is a serious com- Genes that regulate skin pigmentation affect the susceptibil-
plication arising from the natural cutaneous response to UV ity of an individual to skin cancer. Other genes, which can be
exposure. Present evidence suggests that it results from the detected before tumours arise, play a role in UVB-induced
generation of T-suppressor cells, which inhibit the effector immunosuppression (reviewed by Streilein156). Using CHS as
cells of Th1 type. It appears that UV-induced immuno- a test of cutaneous immune response, there are distinct
suppression depresses the function of Th1 cells and enhances phenotypes within the human population, UVB-resistant and
the activity of Th2 cells via cytokines (reviewed by Ullrich145). UVB-sensitive (UVB-R and UVB-S).146 This does not corre-
The combination of these events shifts the normal cutaneous late with skin pigmentation as 40% of dark-skinned individ-
immune response away from cell-mediated immunity. In turn, uals display the UVB-S phenotype.157 Providing further
this causes decreased immunity to intracellular pathogens, evidence that UV-induced immunosuppression has a role in
including bacteria and viruses. This loss of cellular immunity skin cancer, is a study by Yoshikawa and Streilein,146 who
may lead to a loss of immunosurveillance to tumours, possi- found that among basal cell and squamous cell carcinoma
bly arising from the direct action of UV on the skin, as it patients, 90% were of the UVB-S phenotype.
appears that individuals who are susceptible to UV-induced Streilein and Bergstresser158 originally demonstrated that
immunosuppression are more susceptible to skin cancer.146 the C3H/HeN and C57Bl strains of mice were UVB-S, while
the BALB/c strain was UVB-R. F1 hybrids of UVB-R and
UVB-S strains were UVB-S, suggesting that this phenotype
UVA-induced immunosuppression
is dominant. C3H/HeJ were found to be UVB-R, despite
Recently the role of UVA in UV-induced immunosuppression differing from C3H/HeN at only the LPS locus, indicating
has become an area of active research. Many of the studies in that polymorphisms at this locus are important in producing
the previous section were performed using lamps with output the phenotype.158 This locus regulates the production of the
in both the UVA and UVB regions. Today, through the use of pro-inflammatory cytokines IL-1, IL-6 and TNF-α.158 The
554 GJ Clydesdale et al.

TNF-α locus is considered important, as there is a 100% ICAM-1 expression in human skin after a 2 or 8 MED dose
correlation between the TNF-αd allele and the UVB-R exposure of UVB. They found that there was no significant
phenotype.159 induction of either adhesin during the 72 h post-UV treat-
Noonan and Hoffman160,161 also investigated the control of ment, although slight upregulation of ICAM-1 occurred on
UVB immunosuppression in a wide variety of mouse strains endothelial cells.
and found a more complex system of genetic control. Three UVB treatment of melanocytes or a melanoma cell line
distinct phenotypes with differing susceptibilities to UV shows a similar effect of UVB on ICAM-1,164 although a
immunosuppression were found. These were classed as variety of cytokines in the same study were found to upregu-
LO (low susceptibility, 50% immunosuppression with late this molecule (IFN-γ, IL-1, IL-6, IL-7 and TNF-β).
9–12.3 kJ/m2 UV) HI (high susceptibility, 50% immuno- ICAM-1 has also been shown to be downregulated on cul-
suppression with 0.7–2.3 kJ/m2 UV) and intermediate pheno- tured monocytes after UV exposure, thereby reducing their
type (intermediate susceptibility, 50% immunosuppression ability to act as accessory cells for T-cell activation.165
with 4.7–6.9 kJ/m2 UV). To relate this to the work of Intracellular adhesion molecule-1 is an important adhe-
Streilein’s group, BALB/c were classified as LO, C3H/HeN sion molecule in the skin as it mediates the binding of T cells
intermediate and C57BL as HI. These phenotypes do not to keratinocytes,166 and is one of the major adhesion mole-
correlate with the MHC haplotype and the findings of cules involved in the interaction of T cells and APC. The
Noonan and Hoffman160,161 disagree with Streilein’s146 obser- effect of UV on keratinocyte expression of ICAM-1 is
vations, as by their definition C3H/HeN and C3H/HeJ strains bi-phasic with an initial downregulation at 24 h followed by
did not differ in their susceptibility, despite the differences at significant induction at later timepoints; this effect was also
the LPS loci. Also, as the TNF-α loci are located within the dose-related.167 Langerhans cells constitutively express this
MHC loci, differences in these genes likewise failed to affect molecule168 and, hence, downregulation of this molecule
the UVB-immunosuppressive phenotype observed. Noonan could affect the ability of LC to stimulate T cells. Tang and
and Hoffman161 did, however, find that UVB-induced Udey169 suggested that doses of UV light, which were suffi-
immunosuppression was controlled by at least three separate cient to cause downregulation of ICAM-1, were ultimately
loci (Uvs1, Uvs2 and Uvs3), one of which was sex-linked, cytotoxic for the LC.
while the other two were autosomal. None of these genes was Alterations in adhesion molecule interaction between
found to be linked to pigment loci. dendritic cells (DC) and T cells do occur after UV exposure
Streilein’s group has now further characterized the loca- of the skin, as shown by the reduced ability of fluorescein
tion of the UVB susceptibility gene to between the Bat5 and isothiocynate (FITC)+ APC from UV-irradiated mice to
H2-D loci on chromosome 17 in the mouse.162 There are a cluster to T cells. Also observed was an increase in the
number of candidate genes within this region and further numbers of CD8+ T cells in the clusters.170 The alteration
analysis will be required to pinpoint a specific gene for this in T-cell subsets may mean different adhesins are active, or
trait. These genes are also present in the human genome and, that LC are expressing different combinations of adhesion
therefore, may be associated with UVB susceptibility in molecules.
humans. From these studies in experimental inbred animals, More recently decreased expression of E and P cadherins
it is clear that the genetic control of the UVB-immunosup- has been shown to be induced in melanocytes following UV
pressive phenotype is complex; this is likely to be even more irradiation,171 which may alter melanoma binding to keratino-
so in the human population. If current progress is maintained cytes. This may increase the metastatic potential of trans-
it may lead to the identification of the gene or genes respon- formed cells and has an inverse relationship with disease
sible for the UVB sensitivity trait being identified. This could progression in melanoma.171
allow the possibility of screening and therapy for susceptible The Fas/Fas ligand (FasL) system mediates T-cell
persons. responses, including peripheral tolerance and the initiation of
apoptosis in activated lymphocytes and DC. Because apop-
tosis is induced in skin after UV exposure, recent studies
Adhesion and co-stimulatory molecules
have investigated the role of these molecules. Leverkus172
A wide variety of adhesion molecules are expressed by cells found that following UV exposure, keratinocytes undergo
of the skin and the blood vessels in the dermis, including UV-induced apoptosis associated with an increased expres-
integrins, selectins, addressins and those of the immuno- sion of Fas; this could be inhibited by the addition of a
globulin super-family. Many of these adhesion molecules Fas-neutralizing antibody. The role of Fas activation in induc-
play a role in inflammatory responses triggered by a variety ing apoptosis after UV exposure is supported by studies by
of factors, such as ultraviolet light. E-Selectin (ELAM-1) is Rehemtulla et al.173 and Aragane et al.174 The latter group
expressed at high levels at sites of acute UVB inflammation found that the transfer of UV-induced suppression was criti-
and acts as a vascular homing molecule for neutrophils and cally dependent on the expression of both Fas and FasL by the
memory T lymphocytes.19 The intracellular adhesion mole- recipient mice that received the suppressor cells; it was
cule (ICAM) and vascular cell adhesion molecule (VCAM) not required on the transferred T suppressor cells.175 Co-
families of adhesion molecules are thought to act in a similar incubation of these cells with DC in vitro was associated with
way during immune responses. enhanced DC apoptosis, which was dependent on Fas and
E-selectin is upregulated on dermal postcapillary venules FasL expression by the DC. This effect could be overcome by
during inflammatory responses,163 including upregulation the addition of IL-12, which was postulated to rescue DC
on vascular endothelium and polymorphonuclear cells after from apoptosis by downregulating Fas expression on DC.175
UV exposure.19 Norris et al.19 also examined VCAM-1 and Hill et al.176 found that FasL expression is not essential for
Ultraviolet light induced injury 555

the transfer of UV-induced immune suppression against cytokine.185 However, treatment with antibodies against cis-
alloantigen, unlike antigens such as C. albicans and FITC. UCA fully restored DTH responses, but did not restore CHS
However, it was essential when the allogeneic cells were responses.23,186 Hart and colleagues found that the use of anti-
disrupted, suggesting that either a different APC population bodies against cis-UCA partially restored the CHS response
was involved or that FasL expression on the intact allogeneic to 2,4,6-trinitrochlorobenzene (TNCB),187 indicating that
cells was responsible for this effect. Immune responses to UV-UCA is playing a major role, but that other mechanisms
alloantigen could not be restored by DNA repair, which also may be involved. Furthermore, they have suggested that mast
suggested that a different pathway is involved, perhaps cells play a critical role in the UCA-mediated suppression of
involving urocanic acid. CHS responses.188
While it appears that the Fas/Fas ligand system is After UV exposure of the skin, mast cells are thought to
extremely important in the production of UV-induced release histamine which, in turn, leads to the production of
immunosuppression, the interactions of cell surface and PG.159 This sequential production of histamine and PG must
adhesion molecules after UV exposure will continue to be be downstream of the photo-isomerization of UCA, as cis-
an important area of study. No doubt, such investigations UCA and histamine effects are not cumulative. Likewise the
will expand to include other adhesion and costimulatory same study demonstrated that the effects of histamine and
molecules. prostanoid antagonists were not cumulative and, thus, pro-
vided further evidence that they were produced sequentially
in this pathway. Additional evidence, although perhaps more
Cytokines and soluble factors produced after cutaneous circumstantial, is that cis-UCA can induce mast-cell degran-
ultraviolet exposure ulation161 and that the extent of UV-induced suppression has
been linked in some mouse strains to mast cell number and
Urocanic acid function.162 However, another group presents evidence that
De Fabo and Noonan177 proposed that urocanic acid (UCA), the suppressive signal arises in the epidermis and that mast
a compound found within the epidermis, was the chro- cells are not an essential requirement, but IL-10 production
mophore for UV-induced immunosuppression. This sub- by epidermal cells appeared to be.117 They found that trans-
stance is produced by the deamination of histidine by the planting UV-irradiated epidermis to the body wall of mast
enzyme histidine ammonia lyase (histidase). The enzyme is cell-deficient mice still generated UV-induced immune
present in high amounts in only two tissues, the skin and suppression. An alternative explanation is that the time
liver.178 Urocanic acid accumulates in the skin due to a lack between UV exposure and the harvesting of the epidermis
of the catabolic enzyme urocanase.178 Urocanic acid absorbs allowed soluble factors, produced by the degranulation of
light in the ultraviolet region, undergoing photoisomerization mast cells, sufficient time to penetrate the epidermal layer.
from the trans to the cis isomer (reviewed by Noonan and De The difference may also be due to differing UV-exposure
Fabo148). regimes, in which the latter study used a series of low-dose
Urocanic acid was proposed as the photoreceptor exposures whereas Hart et al.159 used a single high-dose
mediating UV-induced immunosuppression due to UCA’s exposure. Thus, from recent studies it is clear that mast cells
absorption spectrum closely matching that of the immuno- do play an important role in the generation of immuno-
suppression action spectrum (more so than DNA), together suppression; however, the exact role remains uncertain.
with its superficial location in the stratum corneum and From the accumulating evidence it is clear that UCA has
photochemical properties.177 The administration of UV- a major role in UV-induced immunosuppression of DTH
irradiated urocanic acid (UV-UCA) to mouse skin suppresses responses and a lesser one in CHS responses. However, the
DTH to herpes simplex virus,134 as well as increasing UV- actions of cis-UCA do not exclude DNA damage being
induced tumour yield and malignancy in the hairless important as both UCA and DNA mediate effects associated
mouse.179 In addition cis-UCA impairs induction of CHS and with immunosuppression.189
induces tolerance.180 Cis-UCA treatment also inhibits
alloantigen presentation by LC-enriched epidermal cells in Cytokines
MLR experiments.181 Urocanic acid increases keratinocyte
production of PGE in the presence of histamine and induces Ultraviolet light exposure of the skin induces many changes
weak ICAM-1 expression, which is increased when com- to the skin microenvironment, including the production of
bined with TNF-α.182 cytokines.190–192 In the epidermis the main secretor of cyto-
Ulraviolet-irradiated urocanic acid causes defects in anti- kines after UV exposure are the keratinocytes, and the
gen presentation by splenic DC,183 as well as reducing their changes in the cytokine profile of this cell have been the
numbers, and altering the morphology of LC in mouse skin180 subject of intensive study (reviewed by Takashima and
in a manner similar to the observed effects of cutaneous UVB Bergstresser192). Langerhans cell production of cytokines
treatment. In contrast to these results, topical application of after UV exposure of skin, while undoubtedly important, has
cis-UCA had no effect on the number of DC within the drain- not been extensively investigated. Further studies on many of
ing lymph node.184 The addition of anti-TNF-α antibodies these cytokines are required to define their role in altering the
was able to block the decrease in LC from the epidermis, but skin immune system.
this did not eliminate the changes in morphology observed by
Kurimoto and Streilein.180 Moreover, the suppression of CHS Interleukin-1 The IL-1 system consists of interaction
by UV-UCA appears to be mediated via TNF-α, although the between IL-1α, IL-1β, the IL-1 receptor antagonist (IL-1RA)
induction of tolerance or DTH was not mediated via this and the IL-1 receptors, type 1 and type 2. Exposure of the
556 GJ Clydesdale et al.

skin to UV light may alter the balance of this complex in IL-4-deficient mice has confirmed these findings210
cytokine system. Ansel et al.193 found that exposure of cul- and while IL-4-deficient mice were susceptible to local
tured keratinocytes to UVB results in increased production of immune suppression, they did not develop systemic immune
IL-1 both intra- and extracellularly. UVB exposure of murine suppression.211
skin in vivo caused epidermal cells to produce IL-1. Inter-
leukin-1 production has been detected in the skin, post-UV Interleukin-10 Schwarz et al.197 showed that serum from
exposure in humans.194,195 While LC production of IL-1 UVB-irradiated mice was able to induce systemic suppres-
appears to be unaltered by UV,196 the production of a specific sion. Furthermore, UVB exposure of cultured human
inhibitor of IL-1 is also observed after UV exposure of epi- keratinocytes resulted in the production of IL-10 in vitro,
dermal cells and keratinocytes.197 The inhibitor was termed which caused systemic immunosuppression when the super-
contra-IL-1 and was found in the serum of UVB-irradiated natants were injected into mice.126 Additional evidence of
mice.198 A possible candidate is IL-1RA, which binds to the UV-induced IL-10 production by human keratinocytes was
IL-1 receptor but produces no activation.199 Hirao200 demon- shown both in vitro212,213 and in vivo.212 Grewe et al.213 found
strated that a 100-fold excess of IL-1RA is needed to inhibit that UVB strongly stimulated IL-10 production; however,
the biological function of IL-1 and that in human skin un- UVA-1 (340–400 nm) wavelengths were found to be the most
exposed to UV, IL-1α dominates. However, in UV-exposed stimulatory. In contrast, Jackson et al.214 reported that human
skin IL-1RA is dominant.200 Furthermore, in the same study, keratinocytes were unable to produce IL-10. This was
a UV dose of 2 MED was shown to dramatically upregulate supported by Teunissen et al.,215 who were unable to detect
IL-1RA in the stratum corneum of human volunteers, leading IL-10 secretion from normal or transformed keratinocytes.
to the suggestion that this could inhibit excessive inflamma- Higher doses of UV were required to produce barely
tion and may account for some of the UVB-induced immuno- detectable IL-10 mRNA production with extensive PCR, but
suppression. no protein was detected. In reply Grewe et al.216 suggested
Studies using the HeCat keratinocyte cell line, have that the RT-PCR method used by Jackson may have lacked
demonstrated that TGF-α, TGF-β, IFN-γ, IL-6 and TNF-α the sensitivity of their system and also argued that the use of
increase IL-1α production, while no effect on mRNA of non-neutralizing antibodies in their enzyme-linked immuno-
IL-1RA was evident. This suggests that the balance between assays increased sensitivity.
IL-1α and IL-1RA is due to the levels of IL-1α, not IL-1RA, Although human keratinocyte production of IL-10 is
which remain constant.201 This is in direct contrast to Hirao’s controversial, this may be a minor contributor of epidermal
hypothesis. IL-10, as Kang et al.217 only detected slight increases in
Interestingly, while proposed as antagonistic in inflam- keratinocyte IL-10 mRNA after UVB exposure. They
mation, immunosuppressive effects of both IL-1RA and reported that large amounts of IL-10 were produced by
IL-1α have been demonstrated. IL-1RA injection into the CD11b+ macrophages, which have been shown to infiltrate
skin prior to sensitization with 2,4-dinitrofluorobenzene the epidermis post-UVB exposure.21 This group demon-
(DNFB) inhibits ear swelling by 36–43%.202 IL-1α has been strated that the production of IL-10 by skin-infiltrating
shown to suppress the ability of LC to present tumour anti- macrophages far outweighed that of the keratinocytes,
gens to CD4+ T cells,203 as well as reducing CHS.204 Enk suggesting that infiltrating macrophages were the major
et al.205 demonstrated that LC from IL-1α-injected mice have source of IL-10 in the epidermis after UVB exposure of
reduced capacity to stimulate anti-CD3 treated T cells. It human skin.
appears that IL-1α and IL-1β, which is only produced by LC Interleukin-10 has been shown to downregulate many
in the epidermis, have different roles in CHS responses, with aspects of the skin immune system, including the CHS reac-
IL-1β being crucial in the induction of CHS.205 IL-1β expres- tion.218 Such inhibition of CHS occurred during the effector
sion is increased in LC after contact sensitizer application to phase of CHS and not during the induction of CHS.219
the skin206 but, as yet, no evidence has been forthcoming on However, in the same study, IL-10 inhibited both the initia-
IL-1β production after UV exposure. tion of DTH and its effector phase. This was supported by
In the skin, interactions between IL-1α, IL-1β and IL-1 Rivas and Ullrich,209 who found that the systemic adminis-
receptor type 1 regulate inflammatory reactions; IL-1RA tration of anti-IL-10 antibodies to mice blocked the activity
and IL-1 receptor type 2 appear to downregulate these of UV suppressor cells in DTH, but not the cells that suppress
responses.207 Further studies of the interaction of this system CHS. Ullrich’s group140 has also demonstrated that the
after UV exposure of the skin should reveal in detail the role injection of an anti-IL-10 antibody into UV-irradiated,
of the IL-1 family in the suppression of cutaneous immune B. Burgdorferi-immunized animals restored the IgG2a and
responses. IgG2b antibody responses and DTH to normal. This provided
further evidence for the role of IL-10 in altering the balance
Interleukin-4 While there is no evidence for IL-4 produc- of the immune responses after cutaneous UV exposure.
tion by cells of the skin after UV light, IL-4 can be detected Local IL-10 production in the skin, by infiltrating
in lymph nodes draining the irradiated site.208 CD4+ Th2 cells macrophages, restricts the ability of LC to stimulate an
isolated from the spleen of UV-irradiated mice produce effective Th1 cell response. Instead anergy of Th1 cells has
IL-4.143 Furthermore, antibodies to IL-4 are able to reduce been observed, but the ability to stimulate a Th2 response was
the ability of UV-induced suppressor cells to suppress DTH unaltered.220 This is supported by Ullrich221 who found that
responses, but no reduction for CHS responses was adding IL-10 from UV-irradiated cultured keratinocytes to
observed.209 This suggests that the suppression of these splenic APC caused inhibition of antigen presentation to Th1
responses involves different mediators. A more recent study cells and enhanced stimulation of Th2 responses. LC also
Ultraviolet light induced injury 557

lose their ability to stimulate a primary allogeneic response of different cell types in the skin. These include mast cells,232
when incubated with IL-10.222 This may be due to the loss of in which it is stored preformed; it is induced in keratino-
important costimulators such as downregulation of B7-2 that cytes,233 activated LC234 and dermal DC.235 Two TNF-α
occurs after addition of IL-10 to human peripheral blood receptors are described; TR60 (Type I, 55 kDa) and TR80
DC.223 The ability of LC to present tumour antigens is also (type II, 75 kDa), with lymphoid cells expressing mainly type
downregulated by incubation with IL-10, which overcomes II and cells of epithelial origin, type I receptors.236 The
the GM-CSF-induced maturation of normal LC APC func- expression of this cytokine is dramatically increased after
tion. Ullrich145 showed both in vitro and in vivo that adminis- exposure of the skin to UVB irradiation.195 TNF-α is
tration of anti-IL-10 mAb restores the lost APC function of considered to have a major role in UVB-induced changes in
splenic DC. inflammatory and immune reactions, including downregula-
The many downregulatory effects of IL-10 on, for tion of CHS responses.237 Cis-UCA generated by the UV
example, CHS, DTH and APC function of LC, and its ability exposure of the epidermis appears to mediate its effects on
to stimulate Th2 cells while inhibiting Th1 cells has resulted CHS via TNF-α.180
in IL-10 being regarded by some as ‘the primary mediator of Tumour necrosis factor-α affects inflammatory and
UVB-induced local, as well as systemic, suppression’.192 immunological responses in the body. These include the
induction of other cytokines, including IL-1α, but not its
Other Interleukins The production of a number of other receptor antagonist.201 TNF-α also has a role in the recruit-
interleukins occurs after UV-light exposure of the epidermis. ment of cells to areas of inflammation by altering adhesion
While IL-3 is produced in mouse skin after irradiation it molecule expression in the surrounding environment.72 Intra-
does not appear to be generated by human keratinocytes and dermal injection of recombinant human TNF-α upregulates
IL-3-like activity in human skin can be inhibited by anti- ICAM-1, VCAM-1 and E-selectin in vivo.238 E-selectin was
bodies to GM-CSF and IL-6.224 upregulated on dermal microvascular endothelial cells after
Interleukin-6 is a pro-inflammatory cytokine that is release of TNF-α by dermal mast cells.163 CD44 also appears
produced by human keratinocytes;225 the peak serum levels to be upregulated, allowing increased binding of leucocytes
occur 12 h after severe sunburning, correlating with the to the endothelial surfaces.239 The increased expression of
development of fever.226 IL-6 is involved with the production CD44 and E-selectin is involved with increased infiltrates
of acute phase proteins, but its role in the skin immune of neutrophils and mononuclear cells following UV light
system is unknown, although there is some evidence that, exposure.19,72,239
after UVB irradiation, the appearance of IL-10 in plasma Additional evidence of the importance of TNF-α comes
may be IL-6 regulated.227 from the study of Walsh76 who found that UVB irradiation
Interleukin-8 is a pro-inflammatory cytokine produced in in vitro and in vivo resulted in TNF-α release from mast cells.
the epidermis and it causes neutrophil chemotaxis to sites of This, in turn, led to increased ICAM-1 and E-selectin expres-
inflammation. IL-8 production is greatly increased within an sion on endothelium within 2 h of UVB exposure. This was
hour of UV exposure (reviewed by Luger and Schwarz191); independent of the epidermis, suggesting that keratinocytes
the levels of IL-8 mRNA increased 11–13-fold.228 The accu- only make a small contribution to the acute release of
mulation of neutrophils in sunburned skin26,74 correlates with TNF-α. However, due to the extent of the epidermis and the
the production of this cytokine.72 keratinocyte production of this cytokine after UVB irradia-
Interleukin-12 has been demonstrated to be upregulated in tion, it is likely that TNF-α release by keratinocytes
human keratinocytes after UVA, but not UVB, exposure.229 contributes, in particular, to the later stages of the UVB-
IL-10 was not found to be upregulated in this study, provid- inflammatory process.
ing further evidence of the photoprotective effect of UVA put Tumour necrosis factor-α was originally named because
forward by Reeve et al.150 of its ability to kill tumour cell lines. Today it is a cytokine
The induction of IL-15 by keratinocytes and dermal known to be involved with the induction of apoptosis.236
fibroblasts has been noted after UV exposure.230 Blauvelt T-cell mediated TNF-α-induced apoptosis occurs for mature
et al.231 reported that freshly isolated keratinocytes and cul- T cells via the TR-80 receptor. This is more important in
tured keratinocytes expressed IL-15 mRNA in vitro, which mediating CD8+ T-cell apoptosis than CD4+ T-cell apoptosis,
was downregulated by UV exposure in a time- and dose- which is mediated mostly via Fas and Fas L interaction.240
dependent manner. Blauvelt et al.231 were unable to explain The 55 kDa version of the TNF receptor is expressed by
their contrasting results with those of Mohamadzadeh et al.230 keratinocytes and has a ‘death domain’ homologous to that of
Blauvelt et al.231 also showed that IL-15 mRNA was Fas. This has been implicated in the induction of apoptosis in
expressed by LC and blood DC, and proposed that IL-15 may non-lymphoid cell apoptosis.236 TNF-α has recently been
contribute to the enhancement of Th1-like responses. Hence, shown to be involved in the formation of sunburn cells,241 as
downregulation of its production after UVB exposure may the injection of anti-TNF-α antibody reduced UV-induced
contribute to UV-induced reduction of Th1-like responses. sunburn cell formation, although the cytokine by itself was
Before the role of this cytokine can be established in the not sufficient to induce sunburn cells.
immune alterations following UV exposure, further studies Tumour necrosis factor-α has a role in regulating the
are required to determine whether its production is enhanced survival and migration of DC found in the skin. TNF-α
or suppressed by UV light. supports the survival of LC in the murine system, but this did
not result in the acquisition of accessory cell function.233 In
Tumour necrosis factor-α Tumour necrosis factor-α (TNF-α) the ovine immune system, TNF-α supports survival of affer-
is a pro-inflammatory cytokine that is produced by a number ent lymph DC, whereas GM-CSF does not.242
558 GJ Clydesdale et al.

Tumour necrosis factor-α has been proposed to stimulate Transforming growth factor-β
LC migration from the skin to the draining lymph node.243
Transforming growth factor-β (TGF-β), in particular the
When increasing doses of TNF-α were injected intradermally
TGF-β1 isoform, has been shown to mediate inflammatory
into BALB/c mice it was observed that DC accumulation in
and immune effects, some of which have similar characteris-
the lymph node was increased after just 2 h.243 Subsequently
tics to UVB-mediated effects.256 Because human keratino-
it was found that TNF-α was required for both DC migration
cytes have been shown to upregulate the production of this
and optimal contact sensitization.244 In contrast, Yoshikawa237
cytokine 24 h after UV exposure,256 it is likely that further
reported that injection of 50 ng TNF-α (identical in amount
study of this cytokine will contribute to our understanding of
and activity to that used by the previous group) intradermally,
UV-induced immunosuppression.
followed by cutaneously applied hapten, led to the retention
of LC within the epidermis. However, previous studies by
Streilein’s group157 had shown that there was indeed a 30% Other soluble factors
reduction of Ia+ (LC) within the epidermis after 2 h, but LC
The production of other cytokines and soluble factors after
numbers returned to normal within 24 h.157 These data
UV exposure of the skin requires further investigation. These
suggest that the differing time courses of experiments may
include VEGF, basic fibroblast growth factor (bFGF)257 and
have led to variation in the results of these two groups.
nerve growth factor (NGF),258 which provide mitogenic
Further evidence for the DC-depleting effect of TNF-α
stimuli to endothelial cells, keraintocytes, fibroblast and
comes from work, in which systemic administration of the
nerve cells. The production of bFGF together with a decrease
cytokine depleted DC from the heart, kidney and skin.245
in IFN-β is thought to lead to increased angiogenesis and the
More recently it was found using skin organ culture that low
formation of new blood vessels.259 It also appears that neuro-
doses of TNF-α (50–100 U/mL) enhanced LC emigration.246
peptides, such as substance P and calcitonin gene-related
However, high doses (> 1000 U/mL) suppressed emigration,
peptide (CGRP), are increased after cutaneous UV exposure.
possibly due to toxicity.246 At this time it is not known
CGRP can induce mast-cell degranulation and the release of
whether TNF-α acts on DC directly or via other mechanisms
TNF-α. Futhermore, the injection of CGRP into the skin
(e.g. CD40 ligation).247
reduces LC density and impairs CHS induction, while the
addition of CGRP anatagonists can overcome the suppression
of CHS induced by UV exposure. The roles of cytokines and
Prostaglandins
soluble factors are crucial to immunological events following
Increases in prostaglandin (PG) production after UV expo- UV exposure of the skin. It also appears to be multifactorial
sure of the skin have been demonstrated,248 and the and future studies will elaborate further on individual com-
prostaglandins produced are PGE2, PGF2α and PG6-oxo-F1α. ponents of this complicated event.
Treatment of the skin with indomethacin (an inhibitor of PG
synthesis) after UV exposure led to a decrease in inflamma-
tion,249,250 but did not prevent decreases in the density of Effects of ultraviolet on Langerhans cells and antigen
LC.250 This was partially due to indomethacin acting as a sun- presentation
screening agent; however, oral administration did not stop LC
Langerhans cells are the only constituent APC type found
depletion, but did partially restored CHS responses after UV
within the epidermis and after exposure to UV alterations in
exposure.251,252 Interestingly both TNF-α and IL-1α, are pro-
LC morphology and kinetics have been observed.66,67 Alter-
duced in the skin after UV exposure, affecting DC function
ations in LC were first shown by Fan et al.,260 who found they
and upregulating keratinocyte synthesis of PGE2.253 PGE2
were difficult to demonstrate in human epidermis after expo-
interacts with the cytokine cascade including IL-4 and IL-10,
sure to UV light. In 1967 Wolff and Winkelmann261 treated
which are responsible for the UV-induced systemic immune
guinea pig skin with UV light, but no changes in LC distrib-
suppression.254 This more recent finding has highlighted the
ution were observed. While the exposure times were noted,
importance of PGE in the cutaneous UV response.
no treatment dose was given in either study. In contrast to
this, a single exposure of human skin to 6 MED of sunlight,
reported by Zelickson and Mottaz,262 showed that the number
Complement
of LC present in the epidermis decreased. Ultraviolet-
Recent studies have shown that the complement component damaged LC expressing thymine dimers have been shown to
C3 may be involved with the generation of UV immuno- migrate to the lymph node 1 h after UV exposure,263 indicat-
suppression and antigenic tolerance.255 Using a variety of ing that the depletion of LC from the skin after UV irradia-
systems, including C3 knockout mice and the inhibition of tion may be associated with increased cell migration. This
cleavage of C3 to C3b, the photoimmunosuppression to has now been confirmed by significant, dose-dependent
DNFB was reversed and the use of a soluble receptor for increases in the rate of LC migration from sheep skin after
complement reduced the infiltration of CD11b+ macro- exposure to varying doses of UVB.264,265
phages. Furthermore, ligation of the iC3b receptor has a While the depletion of LC from the skin would reduce the
similar effect on UV-induced immunosuppression.255 The immunological ability of the epidermis to respond to foreign
interaction between these two molecules appears to reduce antigens, this is not the only effect UV light has on LC. It has
the production of IL-12, while increasing IL-10, which is been extensively demonstrated that UV irradiation affects
considered to be a crucial step for the generation of immuno- APC function. Levis et al.266 showed that the ability of UV-
suppression. treated APC to stimulate hapten (DNCB)-specific autologous
Ultraviolet light induced injury 559

lymphocytes in primary culture was abrogated, and the ability Ullrich et al.274 was able to inhibit UV-induced suppres-
to stimulate blast cells in secondary culture was diminished, sion by the in vivo addition of mAb to B7-2, but not B7-1.
but not eliminated. The effect of UV light on APC function The antibodies to B7-2 also stopped secretion of IL-10 into
was also studied in vivo by Greene et al.,113 who reported that the plasma, providing a mechanism for the lack of suppres-
TNP-derivatized APC from UV-irradiated mice failed to sion observed in vivo. Any loss of B7 expression and/or
induce an immune response in UV-irradiated syngeneic mice, upregulation following UV exposure of the skin would render
when compared to normal APC. This failure to induce immu- the LC migrating to the lymph node less able to stimulate
nity was associated with suppressor T cells. In vitro studies T cells and, hence, lead to anergy275 and specific downregu-
by Stingl et al.267 have shown that the ability of epidermal lation of Th1 responses.276 In support of this hypothesis is
cells to stimulate T cells was reduced in a dose-dependent crucial evidence presented by Simon et al.,277 which shows
fashion after exposure to UV. No epidermal cell death was that LC from UV-irradiated skin are able to stimulate Th2
observed, however, as LC made up only 3–5% of epidermal cells, but not Th1 cells. LC from UV-treated skin can induce
cells, specific cytotoxic effects on these cells could not be anergy in Th1 cells,123 which are essential for cellular
ruled out, although similar percentages of LC were found in immune responses such as DTH and CHS. This also provides
epidermal cell preparations, both before and after irradiation. a mechanism for generating Th2 ‘suppressor cells’ and reduc-
The loss of cell surface markers was suggested as the most ing or eliminating Th1 ‘effector cells’, both of which are
likely explanation for these events. This suggestion was con- required for the generation of UV immunosuppression.122 The
sistent with data showing that exposure of LC to UV light interaction between the APC and the antigen specific T cell
lowers ATPase activity in vivo.268 Exposure to milder doses of are crucial to any immune process and this interaction is
UV light in vitro (200 J/m2) had little effect on the retention altered after UV exposure, contributing to the antigen-
of Ia (murine MHC II) antigen or the T6 antigens.269 Recent specific suppressive events that occur after such an exposure.
work suggests that no reduction in MHC II expression is
present in FITC+ DC migrating after a dose of 2 kJ/m2.170
The loss of important costimulator expression may, in
Ultraviolet-induced macrophages
turn, provide a partial explanation for the reduction in LC
function observed after UV exposure. This has been shown as While UV irradiation reduces the allostimulatory capacity of
a reduced T-cell proliferative response267 and by a loss of the DC isolated from the epidermis immediately after treatment,
DTH response.270 Loss of CHS response after UV irradiation Cooper and colleagues found cells isolated from skin 3 days
is also associated with deficient antigen presentation.178 later had an enhanced allostimulatory capacity.271 This
It should also be noted that splenic APC function is down- increased allostimulatory activity was due to T6– DC, which
regulated after UV exposure1 perhaps mediated by cis- had infiltrated the epidermis at this later time.271 These infil-
UCA,183 suggesting a systemic reduction in APC function trating DC expressed the monocyte specific marker, OKM5
after UV exposure of the skin. Locally, the physical damage (CD36),271 suggesting that these cells were of the mono-
of the migrating DC may be responsible for the LC loss of cyte/macrophage lineage. They had the ability to induce
function, as LC migrating from UVB-irradiated sheep skin T-cell proliferation and were able to act as stimulators of an
have a profound loss of dendritic processes compared to LC autologous mixed epidermal cell lymphocyte reaction,271 and
migrating from normal skin.264 It is likely these physical were subsequently shown to express MHC II.17
defects would reduce the ability of LC to act as APC. A later study by Cooper et al.21 showed that a subpopula-
The ability of enriched T6+ LC to effectively induce tion of the infiltrating cells were non-dendritic neutrophils,
a mixed epidermal cell lymphocyte reaction is reduced which is consistent with data obtained from studies on rats,
immediately after exposure of the skin to UV, compared to rabbits, guinea pigs26,32 and humans.74 Another subpopulation
non-irradiated controls.271 This reduction in allostimulatory of infiltrating cells were dendritic, and expressed high levels
activity was not reversed by the addition of indomethacin or of the macrophage marker CD11b, and coexpressed high
IL-1 to the culture system. It can be speculated that the levels of MHC II. Timing of the peak infiltration of this sub-
production of IL-10, by keratinocytes or UV-induced population coincided with an increase in post-UVB epider-
macrophages, may be able to downregulate the costimulator mal APC function from 21% to 59%, a time when LC
B7-2 on LC, as has been demonstrated after addition of depletion was maximal. Hammerberg et al.278 found that this
IL-10 to human peripheral blood DC.223 IL-10 is proposed to subpopulation of infiltrating cells, when isolated from UVB-
mediate the systemic suppression of APC function observed exposed, hapten-treated mouse skin, led to re-sensitization
after UVB exposure.145 Recent evidence supports this argu- rather than tolerance. These cells were also shown to stimu-
ment.272 LC can be induced to express the B7-1 and B7-2 late suppressor inducer cells (Th2) and CD8 cells in the pres-
costimulatory molecules in vitro. However, UVB exposure of ence of IL-2.279,280
epidermal cell suspensions, with doses ranging from 100 to Ultraviolet-induced macrophages are a major source of
200 J/m2, reduced functional B7-1 and B7-2 expression in a IL-10 secreted in the epidermis post-irradiation.217 As IL-10
dose-dependent manner. This was partially restored with is a potent downregulator of Th1 (cellular) immune
submitogenic doses of anti-CD28 mAb.273 Because LC could responses,281 this provides a mechanism by which the infil-
not be returned to their full allostimulatory capacity, Weiss trating macrophage population can suppress cellular immune
et al.273 suggest that this may be due to the presence of responses generated by LC. Recent experiments indicate that
an inhibitory cytokine, possible candidates include PGE, infiltrating UV-induced macrophages appear to come from
IL-1, IL-10 or TNF-α, which are produced after UVB the expansion of a subset of dermal macrophages that are
exposure. CD36+ CD11b+CD1–.282
560 GJ Clydesdale et al.

Kurimoto et al.283 have produced data from murine exper- of cellular changes. The trigger(s) for these effects are
iments that contradict Cooper’s theory278 that macrophages thought to be direct DNA damage of cells, or photoisomer-
mediate the downregulation of immune function after UV. ization of trans to cis-UCA and its subsequent interaction
The investigation of Kurimoto et al. used liposomes contain- with cells in the skin. Until now, detailed mechanistic studies
ing dichloromethylene diphosphonate, which selectively kills have largely been performed in animal models rather than in
phagocytic macrophages, and showed that these cells play a humans. Investigations of the human response to UV expo-
role in generating CHS responses in UV-resistant mice, but sure have concentrated on endpoints such as erythema, DTH
have no effect on tolerance induction.283 Although they responses and the development of cutaneous cancer. What is
endeavoured to compare their results to those of Cooper’s now required, are parallel investigations in humans, which
group, their UV irradiation consisted of four treatments, could serve to define the cellular, biochemical and molecular
whereas Cooper’s group used a single dose. No immunohis- events that occur in human skin after UV exposure.
tochemical studies were performed to show that macrophages These alterations induce changes in the cytokine profile of
were depleted from the skin by the dichloromethylene keratinocytes. Many factors are produced after cutaneous UV
diphosphonate, which may be important as Weber-Matthiesen exposure, while others are reduced, with local and systemic
and Strerry284 found that OKM5+ macrophages are non- effects. After an initial increase in NO, histamine and PGE2,
phagocytic, thus, they may not have ingested the liposomes. which leads to increased vascular permeability, the produc-
While clear evidence exists in humans and mice for the tion of pro-inflammatory cytokines such as IL-1, TNF-α,
infiltration of macrophages into the skin after UV, less is IL-6 and IL-8 closely follows together with the activation of
known about what subsequently occurs to these cells. Evi- complement components. This in turn leads to the down-
dence gathered from mice suggests that following UVB stream production of the anti-inflammatory mediators
exposure of the skin, macrophages do appear in the lymph TGF-β, IL-10 and IL-1RA. There are many other factors of
nodes285 and an increased proportion of DC expressing uncertain role, including neuropeptides, chemokines and
CD11b (Mac1) are found binding to CD4 and an increased other cellular growth factors.
proportion of CD8 positive cells.170 After UV exposure, LC APC function is altered with the
The timing of the maximum migration of UV-induced Th1/Th2 balance directed in favour of Th2-cell function. In
macrophages into the skin coincides with the maximal deple- addition, a population of macrophages that secrete IL-10,
tion of LC. As they secrete IL-10217 and stimulate suppressor thereby, suppressing Th1 responses, migrates into the epi-
inducer T cells,279,280 which both result in downregulation of dermis. These events combine, leading to the generation of
Th1 immune responses, it is likely that the infiltrating suppressor T cells, which produce Th2 cytokines, further dis-
macrophages may contribute to suppression of immune rupting cell-mediated immune responses by inhibiting effec-
responses after UV irradiation. tors of the Th1 subtype. The induction of specific suppression
of CHS and DTH responses is evident and is believed to be
mediated by TNF-α and IL-10, respectively. IL-10 suppresses
γδ T-cell changes
the effector phase of both, possibly by inhibiting APC pro-
Ultraviolet-B exposure of murine skin can also completely duction of IL-12 via downregulation of costimulators. These
deplete dendritic epidermal T cells (DETC) from the epider- changes in the cellular arm of the skin immune system would
mis.286,287 In these experiments depletion was sustained and increase susceptibility to viral infection and greatly reduce
Thy1 DETC did not reappear until 14–22 weeks following any effective immune response to tumours, which may arise
exposure of the skin, whereas the influx of LC into the skin in the skin as a result of UV-induced mutation.
was continuous.286 The doses used were four exposures to
700 J/m2 or a single exposure of 1000 J/m2. Chronic exposure, Acknowledgements
such as that used by Alcalay et al.,287 consisting of 8 kJ/m2
three times per week until the mice developed tumours, The authors acknowledge the support of the National Health
resulted in DETC being consistently absent for up to 43 weeks and Medical Research Council of Australia and the Cancer
post-treatment. LC were observed to return to the skin and Council of Tasmania who have funded research projects
infiltrate tumours.287 This suggested to the authors that the related to this review.
tumours persisted because of the lack of Thy1 DETC and their
cytotoxic potential. Love-Schimenti and Kripke288 found that References
DETC incubated in the presence of IL-2 were capable of
1 Kripke ML. Immunological unresponsiveness induced by ultra-
deleting hapten specific autologous T cells. Thus, it is possi-
violet radiation. Immunol. Rev. 1984; 80: 87–102.
ble that loss of these cells from murine skin may play an
2 de Gruijl FR. Photobiology of photocarcinogenesis. Photochem.
important role in UV-induced carcinogenesis. This observa- Photobiol. 1996; 63: 372–5.
tion is limited to animal species that have a network of DETC 3 Lloyd SA. Stratospheric ozone depletion. Lancet 1993; 342:
in the skin; the lack of such a network in human skin suggests 1156–8.
that they are not important in the human defence system. 4 Chapman RS, Cooper KD, De Fabo EC et al. Solar ultraviolet
radiation and the risk of infectious disease: summary of a work-
Conclusions: Immunity is compromised by cutaneous shop. Photochem. Photobiol. 1995; 61: 223–47.
ultraviolet exposure 5 Green A, Battistutta D, Hart V, Leslie D, Weedon D. Skin cancer
in a subtropical Australian population: incidence and lack of
Human skin is often exposed to UV light in sunlight. This association with occupation. The Nambour Study Group. Am. J.
downregulates the skin immune system by inducing a number Epidemiol. 1996; 144: 1034–40.
Ultraviolet light induced injury 561

6 Osterberg RE, Szarfman A. Assessment of risk for photo- not preventing dendritic cell accumulation in lymph nodes
carcinogenesis: regulatory reviewer viewpoint. Photochem. draining the site of irradiation and contact hypersensitivity
Photobiol. 1996; 63: 362–5. responses. J. Invest. Dermatol. 1995; 105: 264–8.
7 Anderson TF. Light Sources in Clinical Photomedicine. In: Lim 24 Finsen NR. Ueber die Beteutung der Chemiscen Strahlen Des
NA (ed.). Clinical Photomedicice. New York: Marcel Dekker Lichts Fur Medicin und Biologie. Leipzig: Vogel, 1899.
Inc., 1993; 37–58. 25 Partington MW. The vascular response of the skin to ultraviolet
8 Stern WK, Urbach F. The diagnostic significance of the minimal light. Clin. Sci. 1954; 13: 425.
erythema dose. Arch. Dermatol. 1972; 105: 387–93. 26 Logan G, Wilhelm DL. Ultraviolet injury as an experimental
9 Hurks HM, Out-Luiting C, Vermeer BJ, Claas FH, Mommaas AM. model of the inflammatory reaction. Nature 1963; 198: 968–9.
The action spectra for UV-induced suppression of MLR and 27 Diffey BL, Oakley AM. The onset of ultraviolet erythema. Br. J.
MECLR show that immunosuppression is mediated by DNA Dermatol. 1987; 116: 183–7.
damage. Photochem. Photobiol. 1995; 62: 449–53. 28 Farr PM, Diffey BL. Quantitative studies on cutaneous erythema
10 Roberts LK, Beasley DG, Learn DB, Giddens LD, Beard J, induced by ultraviolet radiation. Br. J. Dermatol. 1984; 111:
Stanfield JW. Ultraviolet spectral energy differences affect the 673–82.
ability of sunscreen lotions to prevent ultraviolet-radiation- 29 Owens DW, Knox JM, Hudson HT, Troll D. Influence of wind on
induced immunosuppression. Photochem. Photobiol. 1996; 63: ultraviolet injury. Arch. Dermatol. 1974; 109: 200–1.
874–84. 30 Owens DW, Knox JM, Hudson HT, Troll D. Influence of humid-
11 Cooper KD. Cell-mediated immunosuppressive mechanisms ity on ultraviolet injury. J. Invest. Dermatol. 1975; 64: 250–2.
induced by UV radiation. Photochem. Photobiol. 1996; 63: 31 Logan G, Wilhelm DL. Vascular permeability changes in
400–6. inflammation. I. The role of endogenous permeability factors
12 Eaglstein WH, Sakai M, Mizuno N. Ultraviolet radiation- in ultraviolet injury. Br. J. Exp. Pathol. 1966; 47: 300–14.
induced inflammation and leukocytes. J. Invest. Dermatol. 1979; 32 Logan G, Wilhelm DL. The inflammatory reaction in ultraviolet
72: 59–63. injury. Br. J. Exp. Pathol. 1966; 47: 286–99.
13 Morison WL, Parrish JA, Woehler ME, Krugler JI, Bloch KJ. 33 Logan G, Wilhelm DL. Vascular permeability changes in
Influence of PUVA and UVB radiation on delayed hypersensi- inflammation. II. The effect of lecithinase antagonists in ultra-
tivity in the guinea pig. J. Invest. Dermatol. 1981; 76: 484–8. violet injury in the guinea-pig. Br. J. Exp. Pathol. 1966; 47:
14 Cornelius LA, Sepp N, Li LJ et al. Selective upregulation of 324–30.
intercellular adhesion molecule (ICAM-1) by ultraviolet B in 34 Bachem A. Time factors of erythema and pigmentation pro-
human dermal microvascular endothelial cells. J. Invest. Der- duced by ultraviolet rays of different wavelengths. J. Invest. Der-
matol. 1994; 103: 23–8. matol. 1955; 25: 215.
15 Gilchrest BA, Soter NA, Hawk JL et al. Histologic changes 35 Cotran RS, Pathak MA. The pattern of vascular leakage induced
associated with ultraviolet A-induced erythema in normal by monochromatic UV irradiation in rats, guinea pigs and hair-
human skin. J. Am. Acad. Dermatol. 1983; 9: 213–19. less mice. J. Invest. Dermatol. 1968; 51: 155–64.
16 Kumakiri M, Hashimoto K, Willis I. Biologic changes due to 36 Ramsay CA, Cripps DJ. Cutaneous arteriolar dilatation elicited
long-wave ultraviolet irradiation on human skin: ultrastructural by ultraviolet irradiation. J. Invest. Dermatol. 1970; 54: 332–7.
study. J. Invest. Dermatol. 1977; 69: 392–400. 37 Ramsay CA, Challoner AV. Vascular changes in human skin after
17 Baadsgaard O, Cooper KD, Lisby S, Wulf HC, Wantzin GL. ultraviolet irradiation. Br. J. Dermatol. 1976; 94: 487–93.
Dose response and time course for induction of T6- DR+ human 38 Warin AP. The ultraviolet erythemas in man. Br. J. Dermatol.
epidermal antigen-presenting cells by in vivo ultraviolet A, B, 1978; 98: 473–7.
and C irradiation. J. Am. Acad. Dermatol. 1987; 17: 792–800. 39 Soffen GA, Blum HF. Quantitative measurements of changes in
18 Everett MA, Yeargers E, Sayre RM, Olson RL. Penetration of mouse skin following a single exposure of ultraviolet light.
epidermis by ultraviolet rays. Photochem. Photobiol. 1966; 5: J. Cell Compar. Physiol. 1961; 58: 81–96.
533–42. 40 Rosario R, Mark GJ, Parrish JA, Mihm Jr MC. Histological
19 Norris P, Poston RN, Thomas DS, Thornhill M, Hawk J, changes produced in skin by equally erythemogenic doses of
Haskard DO. The expression of endothelial leukocyte adhe- UV-A, UV-B, UV-C and UV-A with psoralens. Br. J. Dermatol.
sion molecule-1 (ELAM-1), intercellular adhesion molecule-1 1979; 101: 299–308.
(ICAM-1), and vascular cell adhesion molecule-1 (VCAM-1) in 41 Pearse AD, Gaskell SA, Marks R. Epidermal changes in human
experimental cutaneous inflammation: a comparison of ultra- skin following irradiation with either UVB or UVA. J. Invest.
violet B erythema and delayed hypersensitivity. J. Invest. Dermatol. 1987; 88: 83–7.
Dermatol. 1991; 96: 763–70. 42 Ouhtit A, Muller HK, Davis DW, Ullrich SE, McConkey D,
20 Kaidbey KH, Kurban AK. The influence of corticosteroids and Ananthaswamy HN. Temporal events in skin injury and the early
topical indomethacin on sunburn erythema. J. Invest. Dermatol. adaptive responses in ultraviolet-irradiated mouse skin. Am. J.
1976; 66: 153–6. Pathol. 2000; 156: 201–7.
21 Cooper KD, Oberhelman L, Hamilton TA et al. UV exposure 43 Haratake A, Uchida Y, Schmuth M et al. UVB-induced alter-
reduces immunization rates and promotes tolerance to epicuta- ations in permeability barrier function: roles for epidermal
neous antigens in humans: relationship to dose, CD1a-DR+ hyperproliferation and thymocyte-mediated response. J. Invest.
epidermal macrophage induction, and Langerhans cell deple- Dermatol. 1997; 108: 769–75.
tion. Proc. Natl Acad. Sci. USA 1992; 89: 8497–501. 44 Holleran WM, Uchida Y, Halkier-Sorensen L et al. Structural
22 Gilchrest BA, Soter NA, Stoff JS, Mihm Jr MC. The human and biochemical basis for the UVB-induced alterations in
sunburn reaction: histologic and biochemical studies. J. Am. epidermal barrier function. Photodermatol. Photoimmunol.
Acad. Dermatol. 1981; 5: 411–22. Photomed. 1997; 13: 117–28.
23 el-Ghorr AA, Norval M. A monoclonal antibody to cis-urocanic 45 Shono S, Imura M, Ota M, Ono S, Toda K. The relationship
acid prevents the ultraviolet-induced changes in Langerhans of skin color, UVB-induced erythema, and melanogenesis.
cells and delayed hypersensitivity responses in mice, although J. Invest. Dermatol. 1985; 84: 265–7.
562 GJ Clydesdale et al.

46 Olson RL, Gaylor J, Everett MA. Skin color, melanin, and 66 Aberer W, Schuler G, Stingl G, Honigsmann H, Wolff K. Ultra-
erythema. Arch. Dermatol. 1973; 108: 541–4. violet light depletes surface markers of Langerhans cells.
47 Woodward DF, Owen DA. Effect of H1- and H2-receptor J. Invest. Dermatol. 1981; 76: 202–10.
antagonists on cutaneous inflammation evoked by histamine 67 Obata M, Tagami H. Alteration in murine epidermal Langerhans
analogues and UV radiation. Eur. J. Pharmacol. 1982; 77: cell population by various UV irradiations: quantitative and
103–12. morphologic studies on the effects of various wavelengths of
48 Greaves MW, Sondergaard J. Pharmacologic agents released in monochromatic radiation on Ia-bearing cells. J. Invest. Derma-
ultraviolet inflammation studied by continuous skin pefusion. tol. 1985; 84: 139–45.
J. Invest. Dermatol. 1970; 54: 365–7. 68 Nix TE, Nordquist RE, Everett MA. Ultrastructural changes
49 Black AK, Greaves MW, Hensby CN, Plummer NA, Warin AP. induced by ultraviolet light in human epidermis: granular
The effects of indomethacin on arachidonic acid and prosta- and transitional cell layers. J. Ultrastruct. Res. 1965; 12:
glandins e2 and f2alpha levels in human skin 24 h after u.v. B 547–73.
and u.v. C irradiation. Br. J. Clin. Pharmacol. 1978; 6: 261–6. 69 Malorni W, Donelli G, Straface E, Santini MT, Paradisi S,
50 Miller WS, Ruderman FR, Smith Jr JG. Aspirin and ultraviolet Giacomoni PU. Both UVA and UVB induce cytoskeleton-
light-induced erythema in man. Arch. Dermatol. 1967; 95: dependent surface blebbing in epidermoid cells. J. Photochem.
357–8. Photobiol. 1994; 26: 265–70.
51 Snyder DS, Eaglstein WH. Topical indomethacin and sunburn. 70 Tyrrell RM, Pidoux M. Action spectra for human skin cells:
Br. J. Dermatol. 1974; 90: 91–3. estimates of the relative cytotoxicity of the middle ultraviolet,
52 Woodward DF, Owen DAA. Quantitative measurement of the near ultraviolet, and violet regions of sunlight on epidermal
vascualr changes produced by UV radiation and carrageenin keratinocytes. Cancer Res. 1987; 47: 1825–9.
using the guinea-pig ear as the site of inflammation. J. Pharma- 71 Willis I, Cylus L. UVA erythema in skin: is it a sunburn?
col. Meth. 1979; 2: 35–42. J. Invest. Dermatol. 1977; 68: 128–9.
53 Morison WL, Paul BS, Parrish JA. The effects of indomethacin 72 Strickland I, Rhodes LE, Flanagan BF, Friedmann PS. TNF-
on long-wave ultraviolet-induced delayed erythema. J. Invest. alpha and IL-8 are upregulated in the epidermis of normal
Dermatol. 1977; 68: 130–3. human skin after UVB exposure: correlation with neutrophil
54 Deliconstantinos G, Villiotou V, Stavrides JC. Release by ultra- accumulation and E-selectin expression. J. Invest. Dermatol.
violet B (u.v. B) radiation of nitric oxide (NO) from human 1997; 108: 763–8.
keratinocytes: a potential role for nitric oxide in erythema 73 Woodcock A, Magnus IA. The sunburn cell in mouse skin: pre-
production. Br. J. Pharmacol. 1995; 114: 1257–65. liminary quantitative studies on its production. Br. J. Dermatol.
55 Freeman SE, Hacham H, Gange RW, Maytum DJ, Sutherland JC, 1976; 95: 459–68.
Sutherland BM. Wavelength dependence of pyrimidine dimer 74 Hawk JL, Murphy GM, Holden CA. The presence of neutrophils
formation in DNA of human skin irradiated in situ with ultra- in human cutaneous ultraviolet-B inflammation. Br. J. Dermatol.
violet light. Proc. Natl Acad. Sci. USA 1989; 86: 5605–9. 1988; 118: 27–30.
56 Ley RD. Photoreactivation of UV-induced pyrimidine dimers 75 Darr D, Fridovich I. Free radicals in cutaneous biology. J. Invest.
and erythema in the marsupial Monodelphis domestica. Proc. Dermatol. 1994; 102: 671–5.
Natl Acad. Sci. USA 1985; 82: 2409–11. 76 Walsh LJ. Ultraviolet B irradiation of skin induces mast cell
57 Wolf P, Yarosh DB, Kripke ML. Effects of sunscreens and a degranulation and release of tumour necrosis factor-alpha.
DNA excision repair enzyme on ultraviolet radiation-induced Immunol. Cell Biol. 1995; 73: 226–33.
inflammation, immune suppression, and cyclobutane pyrimidine 77 Kligman LH, Murphy GF. Ultraviolet B radiation increases
dimer formation in mice. J. Invest. Dermatol. 1993; 101: 523–7. hairless mouse mast cells in a dose- dependent manner and
58 Daniels F, Brophy D, Lobitz WC. Histochemical responses of alters distribution of UV-induced mast cell growth factor.
human skin following ultraviolet irradiation. J. Invest. Dermatol. Photochem. Photobiol. 1996; 63: 123–7.
1961; 37: 351–7. 78 Ely JO, Ross MH. Absorbtion of light by living cells. Nature
59 Wilgram GF, Kidd RL, Krawczyk WS, Cole PL. Sunburn effect 1949; 163: 906.
on keratinosomes. A report with special note on ultraviolet- 79 Mitchell DL, Jen J, Cleaver JE. Sequence specificity of cyclo-
induced dyskeratosis. Arch. Dermatol. 1970; 101: 505–19. butane pyrimidine dimers in DNA treated with solar (ultraviolet
60 Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological B) radiation. Nucl. Acids Res. 1992; 20: 225–9.
phenomenon with wide-ranging implications in tissue kinetics. 80 Lippke JA, Gordon LK, Brash DE, Haseltine WA. Distribution
Br. J. Cancer 1972; 26: 239–57. of UV light-induced damage in a defined sequence of human
61 Olson RL, Everett MA. Epidermal apoptosis: cell deletion by DNA: detection of alkaline-sensitive lesions at pyrimidine
phagocytosis. J. Cutan. Pathol. 1975; 2: 53–7. nucleoside- cytidine sequences. Proc. Natl Acad. Sci. USA 1981;
62 Duvall E, Wyllie AH, Morris RG. Macrophage recognition of 78: 3388–92.
cells undergoing programmed cell death (apoptosis). Immunol- 81 Vink AA, Berg RJ, de Gruijl FR, Lohman PH, Roza L, Baan RA.
ogy 1985; 56: 351–8. Detection of thymine dimers in suprabasal and basal cells of
63 Cooper KD, Duraiswamy N, Hammerberg C et al. Neutrophils, chronically UV-B exposed hairless mice. J. Invest. Dermatol.
differentiated macrophages, and monocyte/macrophage antigen 1993; 100: 795–9.
presenting cells infiltrate murine epidermis after UV injury. 82 Vink AA, Sontag Y, de Gruijl FR, Roza L, Baan RA. Immuno-
J. Invest. Dermatol. 1993; 101: 155–63. chemical detection of cyclobutane thymine dimers in epi-
64 Godar DE, Lucas AD. Spectral dependence of UV-induced dermal Langerhans cells of ultraviolet B-irradiated hairless
immediate and delayed apoptosis: the role of membrane and mice. Photodermatol. Photoimmunol. Photomed. 1994; 10:
DNA damage. Photochem. Photobiol. 1995; 62: 108–13. 8–12.
65 Godar DE. Preprogrammed and programmed cell death mecha- 83 Hart RW, Setlow RB, Woodhead AD. Evidence that pyrimidine
nisms of apoptosis: UV-induced immediate and delayed apopto- dimers in DNA can give rise to tumors. Proc. Natl Acad. Sci.
sis. Photochem. Photobiol. 1996; 63: 825–30. USA 1977; 74: 5574–8.
Ultraviolet light induced injury 563

84 Pierceall WE, Kripke ML, Ananthaswamy HN. N-ras mutation UV-B-induced acute skin effects and systemic immuno-
in ultraviolet radiation-induced murine skin cancers. Cancer suppression. J. Immunol. 2000; 164: 6199–205.
Res. 1992; 52: 3946–51. 103 Anstey A, Marks R, Long C et al. In vivo photoinduction
85 Roddey PK, Garmyn M, Park HY, Bhawan J, Gilchrest BA. of metallothionein in human skin by ultraviolet irradiation.
Ultraviolet irradiation induces c-fos but not c-Ha-ras proto- J. Pathol. 1996; 178: 84–8.
oncogene expression in human epidermis. J. Invest. Dermatol. 104 Hanada K, Baba T, Hashimoto I, Fukui R, Watanabe S. Pos-
1994; 102: 296–9. sible role of cutaneous metallothionein in protection against
86 Simon MM, Aragane Y, Schwarz A, Luger TA, Schwarz T. UVB photo-oxidative stress – epidermal localization and scavenging
light induces nuclear factor kappa B (NF kappa B) activity activity for superoxide and hydroxyl radicals. Photodermatol.
independently from chromosomal DNA damage in cell-free Photoimmunol. Photomed 1992; 9: 209–13.
cytosolic extracts. J. Invest. Dermatol. 1994; 102: 422–7. 105 Ghosh R, Amstad P, Cerutti P. UVB-induced DNA breaks
87 Abeyama K, Eng W, Jester JV et al. A role for NF-kappa interfere with transcriptional induction of c-fos. Mol. Cell. Biol.
B-dependent gene transactivation in sunburn. J. Clin. Invest. 1993; 13: 6992–9.
2000; 105: 1751–9. 106 Muller HK, Bucana C, Kripke ML. Antigen presentation in the
88 Brash DE, Rudolph JA, Simon JA et al. A role for sunlight skin: modulation by u.v. radiation and chemical carcinogens.
in skin cancer: UV-induced p53 mutations in squamous cell Semin. Immunol. 1992; 4: 205–15.
carcinoma. Proc. Natl Acad. Sci. USA 1991; 88: 10 124–8. 107 Hanisko J, Suskind RR. The effect of ultraviolet radiation on
89 Lubbe J, Reichel M, Burg G, Kleihues P. Absence of p53 gene experimental cutaneous sensitization in guinea pigs. J. Invest.
mutations in cutaneous melanoma. J. Invest. Dermatol. 1994; Dermatol. 1963; 40: 183–91.
102: 819–21. 108 Kripke ML. Antigenicity of murine skin tumors induced by
90 Ziegler A, Jonason AS, Leffell DJ et al. Sunburn and p53 in the ultraviolet light. J. Natl Cancer Inst. 1974; 53: 1333–6.
onset of skin cancer. Nature 1994; 372: 773–6. 109 Kripke ML, Fisher MS. Immunologic responses of the
91 Jiang W, Ananthaswamy HN, Muller HK, Kripke ML. p53 autochthonous host against tumors induced by ultraviolet light.
protects against skin cancer induction by UV-B radiation. Adv. Exp. Med. Biol. 1976; 66: 445–9.
Oncogene 1999; 18: 4247–53. 110 Fisher MS, Kripke ML. Further studies on the tumor-specific
92 Muller-Rover S, Rossiter H, Paus R et al. Overexpression suppressor cells induced by ultraviolet radiation. J. Immunol.
of Bcl-2 protects from ultraviolet B-induced apoptosis but 1978; 121: 1139–44.
promotes hair follicle regression and chemotherapy-induced 111 Morison WL, Parrish JA, Bloch KJ, Krugler JI. In vivo effect
alopecia. Am. J. Pathol. 2000; 156: 1395–405. of UV-B on lymphocyte function. Br. J. Dermatol. 1979; 101:
93 Ziegler A, Jonason A, Simon J, Leffell D, Brash DE. Tumor 513–19.
suppressor gene mutations and photocarcinogenesis. Photo- 112 Hersey P, Haran G, Hasic E, Edwards A. Alteration of T cell
chem. Photobiol. 1996; 63: 432–5. subsets and induction of suppressor T cell activity in normal
94 Ananthaswamy HN, Fourtanier A, Evans RL et al. p53 muta- subjects after exposure to sunlight. J. Immunol. 1983; 131:
tions in hairless SKH-hr1 mouse skin tumors induced by a 171–4.
solar simulator. Photochem. Photobiol. 1998; 67: 227–32. 113 Greene MI, Sy MS, Kripke M, Benacerraf B. Impairment of
95 Kripke ML, Cox PA, Alas LG, Yarosh DB. Pyrimidine dimers antigen-presenting cell function by ultraviolet radiation. Proc.
in DNA initiate systemic immunosuppression in UV-irradiated Natl Acad. Sci. USA 1979; 76: 6591–5.
mice. Proc. Natl Acad. Sci. USA 1992; 89: 7516–20. 114 Ullrich SE, Azizi E, Kripke ML. Suppression of the induction
96 Applegate LA, Ley RD, Alcalay J, Kripke ML. Identification of delayed-type hypersensitivity reactions in mice by a single
of the molecular target for the suppression of contact hyper- exposure to ultraviolet radiation. Photochem. Photobiol. 1986;
sensitivity by ultraviolet radiation. J. Exp. Med. 1989; 170: 43: 633–8.
1117–31. 115 Kripke ML, Lofgreen JS, Beard J, Jessup JM, Fisher MS. In
97 Yarosh DB, Yee V. SKH-1 hairless mice repair UV-induced vivo immune responses of mice during carcinogenesis by ultra-
pyrimidine dimers in epidermal DNA. J. Photochem. Photo- violet irradiation. J. Natl Cancer Inst. 1977; 59: 1227–30.
biol. 1990; 7: 173–9. 116 Spellman CW, Anderson WL, Bernhard EJ, Tomasi TB. Sup-
98 Vink AA, Yarosh DB, Kripke ML. Chromophore for UV- pression of antibody responses to topically applied antigens by
induced immunosuppression: DNA. Photochem. Photobiol. ultraviolet light irradiation. Induction of phototolerance. J. Exp.
1996; 63: 383–6. Med. 1984; 160: 1891–900.
99 Wolf P, Cox P, Yarosh DB, Kripke ML. Sunscreens and T4N5 117 Noonan FP, De Fabo EC. Ultraviolet-B dose-response curves
liposomes differ in their ability to protect against ultraviolet- for local and systemic immunosuppression are identical.
induced sunburn cell formation, alterations of dendritic epider- Photochem. Photobiol. 1990; 52: 801–10.
mal cells, and local suppression of contact hypersensitivity. 118 Kurimoto I, Kitazawa T, Streilein JW. Studies of delayed sys-
J. Invest. Dermatol. 1995; 104: 287–92. temic effects of ultraviolet B radiation (UVR) on the induction
100 Cruz Jr PD, Leverkus M, Dougherty I et al. Thymidine di- of contact hypersensitivity, 2. Evidence that interleukin-10
nucleotides inhibit contact hypersensitivity and activate the from UVR-treated epidermis is the critical mediator. Immunol-
gene for tumor necrosis factor alpha1. J. Invest. Dermatol. ogy 2000; 99: 134–40.
2000; 114: 253–8. 119 Kitazawa T, Streilein JW. Studies on delayed systemic effects
101 Berg RJ, Ruven HJ, Sands AT, de Gruijl FR, Mullenders LH. of ultraviolet B radiation on the induction of contact hyper-
Defective global genome repair in XPC mice is associated sensitivity, 3. Dendritic cells from secondary lymphoid organs
with skin cancer susceptibility but not with sensitivity to UVB are deficient in interleukin-12 production and capacity to
induced erythema and edema. J. Invest. Dermatol. 1998; 110: promote activation and differentiation of T helper type 1 cells.
405–9. Immunology 2000; 99: 296–304.
102 Garssen J, van Steeg H, de Gruijl F et al. Transcription- 120 Elmets CA, Bergstresser PR, Tigelaar RE, Wood PJ, Streilein
coupled and global genome repair differentially influence JW. Analysis of the mechanism of unresponsiveness produced
564 GJ Clydesdale et al.

by haptens painted on skin exposed to low dose ultraviolet 138 Yasumoto S, Hayashi Y, Aurelian L. Immunity to herpes
radiation. J. Exp. Med. 1983; 158: 781–94. simplex virus type 2. Suppression of virus-induced immune
121 Ullrich SE, Kripke ML. Mechanisms in the suppression of responses in ultraviolet B-irradiated mice. J. Immunol. 1987;
tumor rejection produced in mice by repeated UV irradiation. 139: 2788–93.
J. Immunol. 1984; 133: 2786–90. 139 Rooney JF, Bryson Y, Mannix ML et al. Prevention of ultra-
122 Glass MJ, Bergstresser PR, Tigelaar RE, Streilein JW. UVB violet-light-induced herpes labialis by sunscreen. Lancet 1991;
radiation and DNFB skin painting induce suppressor cells 338: 1419–22.
universally in mice. J. Invest. Dermatol. 1990; 94: 273–8. 140 Brown EL, Rivas JM, Ullrich SE, Young CR, Norris SJ,
123 Simon JC, Tigelaar RE, Bergstresser PR, Edelbaum D, Cruz Kripke ML. Modulation of immunity to Borrelia burgdorferi by
Jr PD. Ultraviolet B radiation converts Langerhans cells from ultraviolet irradiation: differential effect on Th1 and Th2
immunogenic to tolerogenic antigen-presenting cells. Induction immune responses. Eur. J. Immunol. 1995; 25: 3017–22.
of specific clonal anergy in CD4+ T helper 1 cells. J. Immunol. 141 Goettsch W, Garssen J, Deijns A, de Gruijl FR, van Loveren H.
1991; 146: 485–91. UV-B exposure impairs resistance to infection by Trichinella
124 Simon JC, Mosmann T, Edelbaum D, Schopf E, Bergstresser PR, spiralis. Environ. Health Perspect. 1994; 102: 298–301.
Crux Jr PD. In vivo evidence that ultraviolet B-induced 142 Goettsch W, Garssen J, De Gruijl FR, Van Loveren H. UVB-
suppression of allergic contact sensitivity is associated with induced decreased resistance to Trichinella spiralis in the rat is
functional inactivation of Th1 cells. Photodermatol. Photo- related to impaired cellular immunity. Photochem. Photobiol.
immunol. Photomed. 1994; 10: 206–11. 1996; 64: 581–5.
125 Simon JC, Krutmann J, Elmets CA, Bergstresser PR, Cruz Jr PD. 143 Yagi H, Tokura Y, Wakita H, Furukawa F, Takigawa M. TCRV
Ultraviolet B-irradiated antigen-presenting cells display altered beta 7+ Th2 cells mediate UVB-induced suppression of murine
accessory signaling for T-cell activation: relevance to immune contact photosensitivity by releasing IL-10. J. Immunol. 1996;
responses initiated in skin. J. Invest. Dermatol. 1992; 98: 66S–69S. 156: 1824–31.
126 Rivas JM, Ullrich SE. Systemic suppression of delayed-type 144 Moodycliffe AM, Nghiem D, Clydesdale G, Ullrich SE.
hypersensitivity by supernatants from UV-irradiated ker- Immune suppression and skin cancer development: regulation
atinocytes. An essential role for keratinocyte-derived IL-10. by NK T cells. Nat. Immunol. 2000; 1: 521–5.
J. Immunol. 1992; 149: 3865–71. 145 Ullrich SE. Does exposure to UV radiation induce a shift to a
127 Jeevan A, Kripke ML. Alteration of the immune response to Th-2-like immune reaction? Photochem. Photobiol. 1996; 64:
Mycobacterium bovis BCG in mice exposed chronically to low 254–8.
doses of UV radiation. Cell. Immunol. 1990; 130: 32–41. 146 Yoshikawa T, Streilein JW. Genetic basis of the effects of
128 Cestari TF, Kripke ML, Baptista PL, Bakos L, Bucana CD. ultraviolet light B on cutaneous immunity. Evidence that poly-
Ultraviolet radiation decreases the granulomatous response to morphism at the Tnfa and Lps loci governs susceptibility.
lepromin in humans. J. Invest. Dermatol. 1995; 105: 8–13. Immunogenetics 1990; 32: 398–405.
129 Goettsch W, Garssen J, de Klerk A, Herremans TM, Dortant P, 147 De Fabo EC, Kripke ML. Wavelength dependence and dose-
de Gruijl FR, Van Loveren H. Effects of ultraviolet-B exposure rate independence of UV radiation-induced immunologic
on the resistance to Listeria monocytogenes in the rat. Photo- unresponsiveness of mice to a UV-induced fibrosarcoma.
chem. Photobiol. 1996; 63: 672–9. Photochem. Photobiol. 1980; 32: 183–8.
130 Denkins Y, Fidler IJ, Kripke ML. Exposure of mice to UV-B 148 Noonan FP, De Fabo EC. Immunosuppression by ultraviolet B
radiation suppresses delayed hypersensitivity to Candida radiation: initiation by urocanic acid. Immunol. Today 1992; 13:
albicans. Photochem. Photobiol. 1989; 49: 615–9. 250–4.
131 Brozek CM, Shopp GM, Ryan SL et al. In vivo exposure to 149 Skov L, Hansen H, Barker JN, Simon JC, Baadsgaard O.
ultraviolet radiation enhances pathogenic effects of murine Contrasting effects of ultraviolet-A and ultraviolet-B exposure
leukemia virus, LP-BM5, in murine acquired immunodefi- on induction of contact sensitivity in human skin. Clin. Exp.
ciency syndrome. Photochem. Photobiol. 1992; 56: 287–95. Immunol. 1997; 107: 585–8.
132 Goettsch W, Garssen J, De Gruijl FR, Van Loveren H. Effects 150 Reeve VE, Bosnic M, Boehm-Wilcox C, Nishimura N, Ley RD.
of UV-B on the resistance against infectious diseases. Toxicol. Ultraviolet A radiation (320–400 nm) protects hairless mice
Lett. 1994; 72: 359–63. from immunosuppression induced by ultraviolet B radiation
133 Garssen J, Van der Vliet H, De Klerk A et al. A rat cyto- (280–320 nm) or cis-urocanic acid. Int. Arch. Allergy Immunol.
megalovirus infection model as a tool for immunotoxicity 1998; 115: 316–22.
testing. Eur. J. Pharmacol. 1995; 292: 223–31. 151 Reeve VE, Bosnic M, Nishimura N. Interferon-gamma is
134 Ross JA, Howie SE, Norval M, Maingay J, Simpson TJ. Ultra- involved in photoimmunoprotection by UVA (320–400 nm)
violet-irradiated urocanic acid suppresses delayed-type hyper- radiation in mice. J. Invest. Dermatol. 1999; 112: 945–50.
sensitivity to herpes simplex virus in mice. J. Invest. Dermatol. 152 Reeve VE, Tyrrell RM. Heme oxygenase induction mediates
1986; 87: 630–3. the photoimmunoprotective activity of UVA radiation in the
135 Howie S, Norval M, Maingay J. Exposure to low-dose ultra- mouse. Proc. Natl Acad. Sci. USA 1999; 96: 9317–21.
violet radiation suppresses delayed-type hypersensitivity to 153 Skov L, Hansen H, Allen M et al. Contrasting effects of ultra-
herpes simplex virus in mice. J. Invest. Dermatol. 1986; 86: violet A1 and ultraviolet B exposure on the induction of tumour
125–8. necrosis factor-alpha in human skin [published errata appear in
136 Ross JA, Howie SE, Norval M, Maingay J. Systemic adminis- Br. J Dermatol 1998, May, 138 (5): 928 and 1998, August, 139
tration of urocanic acid generates suppression of the delayed (2): 361]. Br. J Dermatol. 1998; 138: 216–20.
type hypersensitivity response to herpes simplex virus in a 154 Longuet-Perret I, Schmitt D, Viac J. Tumour necrosis factor-
murine model of infection. Photodermatol. 1988; 5: 9–14. alpha is involved in the contrasting effects of ultraviolet B and
137 El-Ghorr AA, Norval M. The effect of UV-B irradiation on ultraviolet A1 radiation on the release by normal human
secondary epidermal infection of mice with herpes simplex keratinocytes of vascular permeability factor. Br. J. Dermatol.
virus type 1. J. Gen. Virol. 1996; 77: 485–91. 1998; 138: 221–4.
Ultraviolet light induced injury 565

155 Damian DL, Barnetson RS, Halliday GM. Low-dose UVA and 172 Leverkus M, Yaar M, Gilchrest BA. Fas/Fas ligand interaction
UVB have different time courses for suppression of contact contributes to UV-induced apoptosis in human keratinocytes.
hypersensitivity to a recall antigen in humans. J. Invest. Der- Exp. Cell. Res. 1997; 232: 255–62.
matol. 1999; 112: 939–44. 173 Rehemtulla A, Hamilton CA, Chinnaiyan AM, Dixit VM.
156 Streilein JW. Immunogenetics of sunlight-induced skin cancer. Ultraviolet radiation-induced apoptosis is mediated by acti-
Photochem. Photobiol. 1996; 63: 422–4. vation of CD-95 (Fas/APO-1). J. Biol. Chem. 1997; 272: 25
157 Vermeer M, Schmieder GJ, Yoshikawa T et al. Effects of 783–6.
ultraviolet B light on cutaneous immune responses of humans 174 Aragane Y, Kulms D, Metze D et al. Ultraviolet light induces
with deeply pigmented skin. J. Invest. Dermatol. 1991; 97: apoptosis via direct activation of CD95 (Fas/APO-1) indepen-
729–34. dently of its ligand CD95L. J. Cell Biol. 1998; 140: 171–82.
158 Streilein JW, Bergstresser PR. Genetic basis of ultraviolet-B 175 Schwarz A, Grabbe S, Grosse-Heitmeyer K et al. Ultraviolet
effects on contact hypersensitivity. Immunogenetics 1988; 27: light-induced immune tolerance is mediated via the Fas/Fas-
252–8. ligand system. J. Immunol. 1998; 160: 4262–70.
159 Vincek V, Kurimoto I, Medema JP, Prieto E, Streilein JW. 176 Hill LL, Shreedhar VK, Kripke ML, Owen-Schaub LB. A crit-
Tumor necrosis factor alpha polymorphism correlates with ical role for Fas ligand in the active suppression of systemic
deleterious effects of ultraviolet B light on cutaneous immu- immune responses by ultraviolet radiation. J. Exp. Med. 1999;
nity. Cancer Res. 1993; 53: 728–32. 189: 1285–94.
160 Noonan FP, Hoffman HA. Susceptibility to immunosuppression 177 De Fabo EC, Noonan FP. Mechanism of immune suppression
by ultraviolet B radiation in the mouse. Immunogenetics 1994; by ultraviolet irradiation in vivo. I. Evidence for the existence
39: 29–39. of a unique photoreceptor in skin and its role in photo-
161 Noonan FP, Hoffman HA. Control of UVB immunosuppression immunology. J. Exp. Med. 1983; 158: 84–98.
in the mouse by autosomal and sex-linked genes. Immuno- 178 Noonan FP, Kripke ML, Pedersen GM, Greene MI. Suppression
genetics 1994; 40: 247–56. of contact hypersensitivity in mice by ultraviolet irradiation is
162 Handel-Fernandez ME, Kurimoto I, Streilein JW, Vincek V. associated with defective antigen presentation. Immunology
Genetic mapping and physical cloning of UVB susceptibility 1981; 43: 527–33.
region in mice. J. Invest. Dermatol. 1999; 113: 224–9. 179 Reeve VE, Greenoak GE, Canfield PJ, Boehm-Wilcox C,
163 Waldorf HA, Walsh LJ, Schechter NM, Murphy GF. Early Gallagher CH. Topical urocanic acid enhances UV-induced
cellular events in evolving cutaneous delayed hypersensitivity tumour yield and malignancy in the hairless mouse. Photo-
in humans. Am. J. Pathol. 1991; 138: 477–86. chem. Photobiol. 1989; 49: 459–64.
164 Kirnbauer R, Charvat B, Schauer E et al. Modulation of inter- 180 Kurimoto I, Streilein JW. Deleterious effects of cis-urocanic
cellular adhesion molecule-1 expression on human melano- acid and UVB radiation on Langerhans cells and on induction
cytes and melanoma cells: evidence for a regulatory role of of contact hypersensitivity are mediated by tumor necrosis
IL-6, IL-7, TNF beta, and UVB light. J. Invest. Dermatol. 1992; factor-alpha. J. Invest. Dermatol. 1992; 99: 69S–70S.
98: 320–6. 181 Beissert S, Ullrich SE, Hosoi J, Granstein RD. Supernatants
165 Krutmann J, Khan IU, Wallis RS et al. Cell membrane is a from UVB radiation-exposed keratinocytes inhibit Langerhans
major locus for ultraviolet B-induced alterations in accessory cell presentation of tumor-associated antigens via IL-10
cells. J. Clin. Invest. 1990; 85: 1529–36. content. J. Leukoc. Biol. 1995; 58: 234–40.
166 Dustin ML, Singer KH, Tuck DT, Springer TA. Adhesion of 182 Mitra RS, Shimizu Y, Nickoloff BJ. Histamine and cis-urocanic
T lymphoblasts to epidermal keratinocytes is regulated by acid augment tumor necrosis factor-alpha mediated induction
interferon gamma and is mediated by intercellular adhesion of keratinocyte intercellular adhesion molecule-1 expression.
molecule 1 (ICAM-1). J. Exp. Med. 1988; 167: 1323–40. J. Cell. Physiol. 1993; 156: 348–57.
167 Norris DA, Lyons MB, Middleton MH, Yohn JJ, Kashihara- 183 Noonan FP, De Fabo EC, Morrison H. Cis-urocanic acid, a
Sawami M. Ultraviolet radiation can either suppress or induce product formed by ultraviolet B irradiation of the skin, initiates
expression of intercellular adhesion molecule 1 (ICAM-1) on an antigen presentation defect in splenic dendritic cells in vivo.
the surface of cultured human keratinocytes. J. Invest. Derma- J. Invest. Dermatol. 1988; 90: 92–9.
tol. 1990; 95: 132–8. 184 Moodycliffe AM, Kimber I, Norval M. The effect of ultra-
168 De Panfilis G, Soligo D, Manara GC, Ferrari C, Torresani C. violet B irradiation and urocanic acid isomers on dendritic cell
Adhesion molecules on the plasma membrane of epidermal migration. Immunology 1992; 77: 394–9.
cells. I. Human resting Langerhans cells express two members 185 Shimizu T, Streilein JW. Evidence that ultraviolet B radiation
of the adherence-promoting CD11/CD18 family, namely, induces tolerance and impairs induction of contact hypersensi-
H-Mac-1 (CD11b/CD18) and gp 150,95 (CD11c/CD18). tivity by different mechanisms. Immunology 1994; 82: 140–8.
J. Invest. Dermatol. 1989; 93: 60–9. 186 Moodycliffe AM, Bucana CD, Kripke ML, Norval M, Ullrich
169 Tang A, Udey MC. Inhibition of epidermal Langerhans cell SE. Differential effects of a monoclonal antibody to cis-
function by low dose ultraviolet B radiation. Ultraviolet B urocanic acid on the suppression of delayed and contact hyper-
radiation selectively modulates ICAM-1 (CD54) expression sensitivity following ultraviolet irradiation. J. Immunol. 1996;
by murine Langerhans cells. J. Immunol. 1991; 146: 3347–55. 157: 2891–9.
170 Muller HK, Bucana CD, Kripke ML, Cox PA, Saijo S, 187 Hart PH, Jaksic A, Swift G, Norval M, el-Ghorr AA, Finlay-
Strickland FM. Ultraviolet irradiation of murine skin alters Jones JJ. Histamine involvement in UVB- and cis-urocanic
cluster formation between lymph node dendritic cells and acid-induced systemic suppression of contact hypersensitivity
specific T lymphocytes. Cell. Immunol. 1994; 157: 263–76. responses. Immunology 1997; 91: 601–8.
171 Seline PC, Norris DA, Horikawa T, Fujita M, Middleton MH, 188 Hart PH, Grimbaldeston MA, Swift GJ, Hosszu EK, Finlay-
Morelli JG. Expression of E and P-cadherin by melanoma Jones JJ. A critical role for dermal mast cells in cis-urocanic
cells decreases in progressive melanomas and following ultra- acid-induced systemic suppression of contact hypersensitivity
violet radiation. J. Invest. Dermatol. 1996; 106: 1320–4. responses in mice. Photochem. Photobiol. 1999; 70: 807–12.
566 GJ Clydesdale et al.

189 Reeve VE, Ley RD, Reilly WG, Bosnic M. Epidermal urocanic 208 Araneo BA, Dowell T, Moon HB, Daynes RA. Regulation of
acid and suppression of contact hypersensitivity by ultraviolet murine lymphokine production in vivo. Ultraviolet radiation
radiation in Monodelphis domestica. Int. Arch. Allergy exposure depresses IL-2 and enhances IL-4 production by
Immunol. 1996; 109: 266–71. T cells through an IL-1-dependent mechanism. J. Immunol.
190 Ansel J, Perry P, Brown J et al. Cytokine modulation of ker- 1989; 143: 1737–44.
atinocyte cytokines. J. Invest. Dermatol. 1990; 94: 101S–107S. 209 Rivas JM, Ullrich SE. The role of IL-4, IL-10, and TNF-alpha
191 Luger TA, Schwarz T. Evidence for an epidermal cytokine in the immune suppression induced by ultraviolet radiation.
network. J. Invest. Dermatol. 1990; 95: 100S–104S. J. Leukoc. Biol. 1994; 56: 769–75.
192 Takashima A, Bergstresser PR. Impact of UVB radiation on the 210 el-Ghorr AA, Norval M. The role of interleukin-4 in ultraviolet
epidermal cytokine network. Photochem. Photobiol. 1996; 63: B light-induced immunosuppression. Immunology 1997; 92:
397–400. 26–32.
193 Ansel JC, Luger TA, Green I. The effect of in vitro and in vivo 211 Hart PH, Grimbaldeston MA, Jaksic A et al. Ultraviolet B-
UV irradiation on the production of ETAF activity by human induced suppression of immune responses in interleukin-4–/–
and murine keratinocytes. J. Invest. Dermatol. 1983; 81: mice: relationship to dermal mast cells. J. Invest. Dermatol.
519–23. 2000; 114: 508–13.
194 Kupper TS, Chua AO, Flood P, McGuire J, Gubler U. Inter- 212 Enk CD, Sredni D, Blauvelt A, Katz SI. Induction of IL-10 gene
leukin 1 gene expression in cultured human keratinocytes is expression in human keratinocytes by UVB exposure in vivo
augmented by ultraviolet irradiation. J. Clin. Invest. 1987; 80: and in vitro. J. Immunol. 1995; 154: 4851–6.
430–6. 213 Grewe M, Gyufko K, Krutmann J. Interleukin-10 production by
195 Oxholm A, Oxholm P, Staberg B, Bendtzen K. Immunohisto- cultured human keratinocytes: regulation by ultraviolet B and
logical detection of interleukin I-like molecules and tumour ultraviolet A1 radiation. J. Invest. Dermatol. 1995; 104: 3–6.
necrosis factor in human epidermis before and after UVB- 214 Jackson M, Thomson KE, Laker R, Norval M, Hunter JA,
irradiation in vivo. Br. J. Dermatol. 1988; 118: 369–76. McKenzie RC. Lack of induction of IL-10 expression in human
196 Rasanen L, Reunala T. Langerhans cell antigen presentation keratinocytes [letter]. J. Invest. Dermatol. 1996; 106: 1329–31.
and interleukin-1 production in atopic dermatitis. Acta Derm. 215 Teunissen MB, Koomen CW, Jansen J et al. In contrast to their
Venereol. 1989; 144 (Suppl): 64–6. murine counterparts, normal human keratinocytes and human
197 Schwarz T, Urbanska A, Gschnait F, Luger TA. UV-irradiated epidermoid cell lines A431 and HaCaT fail to express IL-10
epidermal cells produce a specific inhibitor of interleukin 1 mRNA and protein. Clin. Exp. Immunol. 1997; 107: 213–23.
activity. J. Immunol. 1987; 138: 1457–63. 216 Grewe M, Duvic M, Aragane M, Schwarz T, Ullrich SE,
198 Schwarz T, Urbanski A, Kirnbauer R, Kock A, Gschnait F, Krutmann J. Lack of induction of IL-10 expression in human
Luger TA. Detection of a specific inhibitor of interleukin 1 keratinocytes. J. Invest. Dermatol. 1996; 106: 1330–1.
in sera of UVB-treated mice. J. Invest. Dermatol. 1988; 91: 217 Kang K, Hammerberg C, Meunier L, Cooper KD. CD11b+
536–40. macrophages that infiltrate human epidermis after in vivo ultra-
199 Dripps DJ, Verderber E, Ng RK, Thompson RC, Eisenberg SP. violet exposure potently produce IL-10 and represent the major
Interleukin-1 (IL-1) receptor antagonist binds to the 80-kDa secretory source of epidermal IL-10 protein. J. Immunol. 1994;
IL-1 receptor but does not initiate IL-1 signal transduction. 153: 5256–64.
J. Biol. Chem. 1991; 266: 20 331–6. 218 Ferguson TA, Dube P, Griffith TS. Regulation of contact hyper-
200 Hirao T, Aoki H, Yoshida T, Sato Y, Kamoda H. Elevation of sensitivity by interleukin 10. J. Exp. Med. 1994; 179:
interleukin 1 receptor antagonist in the stratum corneum of 1597–604.
sun-exposed and ultraviolet B-irradiated human skin. J. Invest. 219 Schwarz A, Grabbe S, Riemann H et al. In vivo effects of inter-
Dermatol. 1996; 106: 1102–7. leukin-10 on contact hypersensitivity and delayed-type hyper-
201 Phillips WG, Feldmann M, Breathnach SM, Brennan FM. sensitivity reactions. J. Invest. Dermatol. 1994; 103: 211–16.
Modulation of the IL-1 cytokine network in keratinocytes by 220 Enk AH, Angeloni VL, Udey MC, Katz SI. Inhibition of
intracellular IL-1 alpha and IL-1 receptor antagonist. Clin. Exp. Langerhans cell antigen-presenting function by IL-10. A role
Immunol. 1995; 101: 177–82. for IL-10 in induction of tolerance. J. Immunol. 1993; 151:
202 Kondo S, Pastore S, Fujisawa H et al. Interleukin-1 receptor 2390–8.
antagonist suppresses contact hypersensitivity. J. Invest. Der- 221 Ullrich SE. Mechanism involved in the systemic suppression
matol. 1995; 105: 334–8. of antigen-presenting cell function by UV irradiation.
203 Grabbe S, Bruvers S, Granstein RD. Interleukin 1 alpha but not Keratinocyte-derived IL-10 modulates antigen-presenting cell
transforming growth factor beta inhibits tumor antigen presen- function of splenic adherent cells. J. Immunol. 1994; 152:
tation by epidermal antigen-presenting cells. J. Invest. Derma- 3410–16.
tol. 1994; 102: 67–73. 222 Peguet-Navarro J, Moulon C, Caux C, Dalbiez-Gauthier C,
204 Robertson B, Gahring L, Newton R, Daynes R. In vivo Banchereau J, Schmitt D. Interleukin-10 inhibits the primary
administration of interleukin 1 to normal mice depresses their allogeneic T cell response to human epidermal Langerhans
capacity to elicit contact hypersensitivity responses: prosta- cells. Eur. J. Immunol. 1994; 24: 884–91.
glandins are involved in this modification of immune function. 223 Buelens C, Willems F, Pierard G, Delvaux A, Velu T, Goldman
J. Invest. Dermatol. 1987; 88: 380–7. M. IL-10 inhibits the primary allogeneic T cell response to
205 Enk AH, Angeloni VL, Udey MC, Katz SI. An essential role for human peripheral blood dendritic cells. Adv. Exp. Med. Biol.
Langerhans cell-derived IL-1 beta in the initiation of primary 1995; 378: 363–5.
immune responses in skin. J. Immunol. 1993; 150: 3698–704. 224 Kondo S, Ciarletta A, Turner KJ, Sauder DN, McKenzie RC.
206 Enk AH, Katz SI. Early events in the induction phase of contact Failure to detect interleukin (IL) -3 mRNA or protein in human
sensitivity. J. Invest. Dermatol. 1992; 99: 39S–41S. keratinocytes: antibodies to granulocyte macrophage-colony-
207 Kupper TS, Groves RW. The interleukin-1 axis and cutaneous stimulating factor or IL-6 (but not IL-3) neutralize ‘IL-3′ bio-
inflammation. J. Invest. Dermatol. 1995; 105: 62S–66S. activity. J. Invest. Dermatol. 1995; 104: 335–9.
Ultraviolet light induced injury 567

225 Kirnbauer R, Kock A, Neuner P et al. Regulation of epidermal 242 Haig DM, Percival A, Mitchell J, Green I, Sargan D. The
cell interleukin-6 production by UV light and corticosteroids. survival and growth of ovine afferent lymph dendritic cells
J. Invest. Dermatol. 1991; 96: 484–9. in culture depends on tumour necrosis factor-alpha and is
226 Urbanski A, Schwarz T, Neuner P et al. Ultraviolet light enhanced by granulocyte-macrophage colony-stimulating
induces increased circulating interleukin-6 in humans. J. Invest. factor but inhibited by interferon-gamma. Vet. Immunol.
Dermatol. 1990; 94: 808–11. Immunopathol. 1995; 45: 221–36.
227 Nishimura N, Tohyama C, Satoh M, Nishimura H, Reeve VE. 243 Cumberbatch M, Kimber I. Dermal tumour necrosis factor-
Defective immune response and severe skin damage following alpha induces dendritic cell migration to draining lymph nodes,
UVB irradiation in interleukin-6-deficient mice. Immunology and possibly provides one stimulus for Langerhans’ cell migra-
1999; 97: 77–83. tion. Immunology 1992; 75: 257–63.
228 Kondo S, Kono T, Sauder DN, McKenzie RC. IL-8 gene 244 Cumberbatch M, Kimber I. Tumour necrosis factor-alpha is
expression and production in human keratinocytes and their required for accumulation of dendritic cells in draining lymph
modulation by UVB. J. Invest. Dermatol. 1993; 101: 690–4. nodes and for optimal contact sensitization. Immunology 1995;
229 Kondo S, Jimbow K. Dose-dependent induction of IL-12 but 84: 31–5.
not IL-10 from human keratinocytes after exposure to ultra- 245 Roake JA, Rao AS, Morris PJ, Larsen CP, Hankins DF,
violet light A. J. Cell. Physiol. 1998; 177: 493–8. Austyn JM. Dendritic cell loss from nonlymphoid tissues after
230 Mohamadzadeh M, Takashima A, Dougherty I, Knop J, systemic administration of lipopolysaccharide, tumor necrosis
Bergstresser PR, Cruz Jr PD. Ultraviolet B radiation up- factor, and interleukin 1. J. Exp. Med. 1995; 181: 2237–47.
regulates the expression of IL-15 in human skin. J. Immunol. 246 Stoitzner P, Zanella M, Ortner U et al. Migration of Langerhans
1995; 155: 4492–6. cells and dermal dendritic cells in skin organ cultures: aug-
231 Blauvelt A, Asada H, Klaus-Kovtun V, Altman DJ, Lucey DR, mentation by TNF-alpha and IL-1beta. J. Leukoc. Biol. 1999;
Katz SI. Interleukin-15 mRNA is expressed by human 66: 462–70.
keratinocytes Langerhans cells, and blood-derived dendritic 247 Moodycliffe AM, Shreedhar V, Ullrich SE et al. CD40–CD40
cells and is downregulated by ultraviolet B radiation. J. Invest. ligand interactions in vivo regulate migration of antigen-
Dermatol. 1996; 106: 1047–52. bearing dendritic cells from the skin to draining lymph nodes.
232 Gordon JR, Galli SJ. Mast cells as a source of both preformed J. Exp. Med. 2000; 191: 2011–20.
and immunologically inducible TNF-alpha/cachectin. Nature 248 Imokawa G, Tejima T. A possible role of prostaglandins in
1990; 346: 274–6. PUVA-induced inflammation: implication by organ cultured
233 Koch F, Heufler C, Kampgen E, Schneeweiss D, Bock G, skin. J. Invest. Dermatol. 1989; 92: 296–300.
Schuler G. Tumor necrosis factor alpha maintains the viability 249 Farr PM, Diffey BL. A quantitative study of the effect of topical
of murine epidermal Langerhans cells in culture, but in contrast indomethacin on cutaneous erythema induced by UVB and
to granulocyte/macrophage colony-stimulating factor, without UVC radiation. Br. J. Dermatol. 1986; 115: 453–66.
inducing their functional maturation. J. Exp. Med. 1990; 171: 250 Lim HW, Thorbecke GJ, Baer RL, Gigli I. Effect of
159–71. indomethacin on alteration of ATPase-positive Langerhans cell
234 Larrick JW, Morhenn V, Chiang YL, Shi T. Activated Langer- density and cutaneous sunburn reaction induced by ultraviolet-
hans cells release tumor necrosis factor. J. Leukoc. Biol. 1989; B radiation. J. Invest. Dermatol. 1983; 81: 455–8.
45: 429–33. 251 Chung HT, Burnham DK, Robertson B, Roberts LK, Daynes RA.
235 Bruynzeel I, Nickoloff BJ, van der Raaij EM, Boorsma DM, Involvement of prostaglandins in the immune alterations
Stoof TJ, Willemze R. Induction of ICAM-1 expression by caused by the exposure of mice to ultraviolet radiation.
epidermal keratinocytes via a paracrine pathway possibly J. Immunol. 1986; 137: 2478–84.
involving dermal dendritic cells. Arch. Dermatol. Res. 1992; 252 June BD, Roberts LK, Cho BH, Robertson B, Daynes RA.
284: 250–2. Parallel recovery of epidermal antigen-presenting cell activity
236 Grell M, Zimmermann G, Hulser D, Pfizenmaier K, Scheurich and contact hypersensitivity responses in mice exposed to
P. TNF receptors TR60 and TR80 can mediate apoptosis via ultraviolet irradiation: the role of a prostaglandin-dependent
induction of distinct signal pathways. J. Immunol. 1994; 153: mechanism. J. Invest. Dermatol. 1988; 90: 311–16.
1963–72. 253 Grewe M, Trefzer U, Ballhorn A, Gyufko K, Henninger H,
237 Yoshikawa T, Kurimoto I, Streilein JW. Tumour necrosis factor- Krutmann J. Analysis of the mechanism of ultraviolet (UV) B
alpha mediates ultraviolet light B-enhanced expression of radiation-induced prostaglandin E2 synthesis by human
contact hypersensitivity. Immunology 1992; 76: 264–71. epidermoid carcinoma cells. J. Invest. Dermatol. 1993; 101:
238 Groves RW, Allen MH, Ross EL, Barker JN, MacDonald DM. 528–31.
Tumour necrosis factor alpha is pro-inflammatory in normal 254 Shreedhar V, Giese T, Sung VW, Ullrich SE. A cytokine cascade
human skin and modulates cutaneous adhesion molecule including prostaglandin E2, IL-4, and IL-10 is responsible for
expression. Br. J. Dermatol. 1995; 132: 345–52. UV-induced systemic immune suppression. J. Immunol. 1998;
239 Osada A, Nakashima H, Furue M, Tamaki K. Up-regulation of 160: 3783–9.
CD44 expression by tumor necrosis factor-alpha is neutralized 255 Hammerberg C, Katiyar SK, Carroll MC, Cooper KD. Acti-
by interleukin-10 in Langerhans cells. J. Invest. Dermatol. vated complement component 3 (C3) is required for ultraviolet
1995; 105: 124–7. induction of immunosuppression and antigenic tolerance.
240 Zheng L, Fisher G, Miller RE, Peschon J, Lynch DH, Lenardo J. Exp. Med. 1998; 187: 1133–8.
MJ. Induction of apoptosis in mature T cells by tumour necro- 256 Lee HS, Kooshesh F, Sauder DN, Kondo S. Modulation of
sis factor. Nature 1995; 377: 348–51. TGF-beta 1 production from human keratinocytes by UVB.
241 Schwarz A, Bhardwaj R., Aragane Y et al. Ultraviolet-B- Exp. Dermatol. 1997; 6: 105–10.
induced apoptosis of keratinocytes: evidence for partial 257 Halaban R, Langdon R, Birchall N et al. Basic fibroblast
involvement of tumor necrosis factor-alpha in the formation of growth factor from human keratinocytes is a natural mitogen
sunburn cells. J. Invest. Dermatol. 1995; 104: 922–7. for melanocytes. J. Cell. Biol. 1988; 107: 1611–19.
568 GJ Clydesdale et al.

258 Tron VA, Coughlin MD, Jang DE, Stanisz J, Sauder DN. costimulatory molecule CD86 interfere with ultraviolet
Expression and modulation of nerve growth factor in murine radiation-induced immune suppression. Immunology 1998; 94:
keratinocytes (PAM 212). J. Clin. Invest. 1990; 85: 1085–9. 417–23.
259 Bielenberg DR, Bucana CD, Sanchez R, Donawho CK, 275 Linsley PS, Ledbetter JA. The role of the CD28 receptor during
Kripke ML, Fidler IJ. Molecular regulation of UVB-induced T cell responses to antigen. Annu. Rev. Immunol. 1993; 11:
cutaneous angiogenesis. J. Invest. Dermatol. 1998; 111: 864–72. 191–212.
260 Fan J, Schoenfeld RJ, Hunter R. A study of epidermal clear 276 Constant SL, Bottomly K. Induction of Th1 and Th2 CD4+
cells with a special referance to their relationship with the cells T cell responses: the alternative approaches. Annu. Rev.
of Langerhans. J. Invest. Dermatol. 1959; 32: 445–9. Immunol. 1997; 15: 297–322.
261 Wolff K, Winkelmann RK. The influence of ultraviolet light on 277 Simon JC, Cruz Jr PD, Bergstresser PR, Tigelaar RE. Low dose
the Langerhans cell population and its hydrolytic enzymes in ultraviolet B-irradiated Langerhans cells preferentially activate
guinea pigs. J. Invest. Dermatol. 1967; 48: 531–9. CD4+ cells of the T helper 2 subset. J. Immunol. 1990; 145:
262 Zelickson AS, Mottaz J. The effect of sunlight on human 2087–91.
epidermis. A quantitative electron microscopic study of 278 Hammerberg C, Duraiswamy N, Cooper KD. Active induction
dendritic cells. Arch. Dermatol. 1970; 101: 312–15. of unresponsiveness (tolerance) to DNFB by in vivo ultraviolet-
263 Sontag Y, Guikers CL, Vink AA et al. Cells with UV-specific exposed epidermal cells is dependent upon infiltrating class II
DNA damage are present in murine lymph nodes after in vivo MHC+ CD11b bright monocytic/macrophagic cells. J.
UV irradiation. J. Invest. Dermatol. 1995; 104: 734–8. Immunol. 1994; 153: 4915–24.
264 Dandie GW, Clydesdale GJ, Jacobs I, Muller HK. Effects of 279 Baadsgaard O, Fox DA, Cooper KD. Human epidermal cells
UV on the migration and function of epidermal antigen from ultraviolet light-exposed skin preferentially activate
presenting cells. Mutat. Res. 1998; 422: 147–54. autoreactive CD4+2H4+ suppressor-inducer lymphocytes and
265 Dandie GW, Clydesdale GJ, Radcliff FJ, Muller HK. Migration CD8+ suppressor/cytotoxic lymphocytes. J. Immunol. 1988;
of Langerhans cells and γδ+ dendritic cells from UVB- 140: 1738–44.
irradiated sheep skin. Immunol. Cell Biol. 2001; 79: 41–8. 280 Baadsgaard O, Salvo B, Mannie A, Dass B, Fox DA, Cooper KD.
266 Levis WR, Lincoln PM, Dattner AM. Effect of ultraviolet light In vivo ultraviolet-exposed human epidermal cells activate
on dinitrochlorobenzene-specific antigen-presenting function. T suppressor cell pathways that involve CD4+CD45RA+
J. Immunol. 1978; 121: 1496–500. suppressor-inducer T cells. J. Immunol. 1990; 145: 2854–61.
267 Stingl G, Gazze-Stingl LA, Aberer W, Wolff K. Antigen 281 Del Prete G, De Carli M, Almerigogna F, Giudizi MG, Biagiotti R,
presentation by murine epidermal Langerhans cells and its Romagnani S. Human IL-10 is produced by both type 1 helper
alteration by ultraviolet B light. J. Immunol. 1981; 127: (Th1) and type 2 helper (Th2) T cell clones and inhibits their
1707–13. antigen-specific proliferation and cytokine production. J.
268 Toews GB, Bergstresser PR, Streilein JW. Epidermal Langer- Immunol. 1993; 150: 353–60.
hans cell density determines whether contact hypersensitivity 282 Meunier L, Bata-Csorgo Z, Cooper KD. In human dermis,
or unresponsiveness follows skin painting with DNFB. J. ultraviolet radiation induces expansion of a CD36+ CD11b+
Immunol. 1980; 124: 445–53. CD1-macrophage subset by infiltration and proliferation;
269 Czernielewski J, Vaigot P, Asselineau D, Prunieras M. In vitro CD1+ Langerhans-like dendritic antigen-presenting cells are
effect of UV radiation on immune function and membrane concomitantly depleted. J. Invest. Dermatol. 1995; 105: 782–8.
markers of human Langerhans cells. J. Invest. Dermatol. 1984; 283 Kurimoto I, van Rooijen N, Dijkstra CD, Streilein JW. Role of
83: 62–5. phagocytic macrophages in induction of contact hypersensitiv-
270 Perry LL, Greene MI. Antigen presentation by epidermal ity and tolerance by hapten applied to normal and ultraviolet
Langerhans cells: loss of function following ultraviolet (UV) B-irradiated skin. Immunology 1994; 83: 281–7.
irradiation in vivo. Clin. Immunol. Immunopathol. 1982; 24: 284 Weber-Matthiesen K, Sterry W. Organization of the mono-
204–19. cyte/macrophage system of normal human skin. J. Invest. Der-
271 Cooper KD, Fox P, Neises G, Katz SI. Effects of ultraviolet matol. 1990; 95: 83–9.
radiation on human epidermal cell alloantigen presentation: 285 Tang JM, Dunner Jr K, Kripke ML, Bucana C. Characteristics
initial depression of Langerhans cell-dependent function is of antigen-presenting cells involved in contact sensitization of
followed by the appearance of T6-Dr+ cells that enhance normal and UV-irradiated mice. J. Invest. Dermatol. 1992; 99:
epidermal alloantigen presentation. J. Immunol. 1985; 134: 20S–22S.
129–37. 286 Aberer W, Romani N, Elbe A, Stingl G. Effects of physico-
272 Rattis FM, Concha M, Dalbiez-Gauthier C, Courtellemont P, chemical agents on murine epidermal Langerhans cells and
Schmitt D, Peguet-Navarro J. Effects of ultraviolet B radiation Thy-1-positive dendritic epidermal cells. J. Immunol. 1986;
on human Langerhans cells: functional alteration of CD86 136: 1210–16.
upregulation and induction of apoptotic cell death. J. Invest. 287 Alcalay J, Craig JN, Kripke ML. Alterations in Langerhans
Dermatol. 1998; 111: 373–9. cells and Thy-1+ dendritic epidermal cells in murine epidermis
273 Weiss JM, Renkl AC, Denfeld RW et al. Low-dose UVB radi- during the evolution of ultraviolet radiation-induced skin
ation perturbs the functional expression of B7.1 and B7.2 cancers. Cancer Res. 1989; 49: 4591–6.
co-stimulatory molecules on human Langerhans cells. Eur. J. 288 Love-Schimenti CD, Kripke ML. Dendritic epidermal T cells
Immunol. 1995; 25: 2858–62. inhibit T cell proliferation and may induce tolerance by cyto-
274 Ullrich SE, Pride MW, Moodycliffe AM. Antibodies to the toxicity. J. Immunol. 1994; 153: 3450–6.

También podría gustarte