Anda di halaman 1dari 13

Photochemistry and Photobiology, 2008, 84: 450–462

Review

Effects of Visible Light on the Skin†


Bassel H. Mahmoud, Camile L. Hexsel, Iltefat H. Hamzavi
and Henry W. Lim*
Multicultural Dermatology Center, Department of Dermatology, Henry Ford Hospital,
Detroit, MI
Received 1 October 2007, accepted 5 December 2007, DOI: 10.1111 ⁄ j.1751-1097.2007.00286.x

ABSTRACT or infrared components. This review will focus on the effects


of visible light on the skin.
Electromagnetic radiation has vast and diverse effects on
human skin. Although photobiologic studies of sunlight date
back to Sir Isaac Newton in 1671, most available studies HISTORY
focus on the UV radiation part of the spectrum. The effects of The earliest study on photobiology was reported in 1671 by Sir
visible light and infrared radiation have not been, until Isaac Newton, who fractionated sunlight into different colors
recently, clearly elucidated. The goal of this review is to of the rainbow using a prism (1). Radiation outside of the
highlight the effects of visible light on the skin. As a result of visible light spectrum was discovered 125 years later by Sir
advances in the understanding of skin optics, and comprehen- William Herschel and by Johann Ritter. In 1800, Herschel (2)
sive studies regarding the absorption spectrum of endogenous found that a thermometer registered a higher temperature
and exogenous skin chromophores, various biologic effects beyond the visible red end of the spectrum than within it, a
have been shown to be exerted by visible light radiation spectrum now known as infrared radiation. In 1801, Ritter
including erythema, pigmentation, thermal damage and free showed a stronger chemical action on silver chloride beyond
radical production. It has also been shown that visible light the visible violet end of the spectrum (3), hence the term UV
can induce indirect DNA damage through the generation of spectrum.
reactive oxygen species. Furthermore, a number of photoder- In 1798, Robert Willan described sensitivity to sunlight
matoses have an action spectrum in the visible light range, under the term eczema solare (4), a condition which was also
even though most of the currently available sunscreens offer, if reported by Rayer (5). Sir Everard Home’s (6) experimental
any, weak protection against visible light. Conversely, because observation in 1820 in England reported that some compo-
of its cutaneous biologic effects, visible light is used for the nents of sunlight, other than heat, affected the skin, which was
treatment of a variety of skin diseases and esthetic conditions confirmed in 1829 by Davy (7), who investigated whether
in the form of lasers, intense pulsed light and photodynamic different rays of the solar spectrum produce different effects.
therapy. Since ancient times, it has been known that electromagnetic
radiation has injurious as well as useful effects; however, the
science of photobiology only started to fully develop in the
INTRODUCTION past few decades. As the ability to produce specific wave-
Studies in photodermatology have focused mainly on the lengths was developed, the biologic implications of these
UV part of the electromagnetic radiation spectrum. It is wavelengths were identified.
known that UV radiation contains sufficient energy that
produces biologic effects in the skin. Until recently, visible
BIOLOGIC EFFECTS OF LIGHT
light (400–700 nm) has been regarded to have no significant
cutaneous photobiologic effect. Recent developments in Electromagnetic radiation is classified based on wavelength.
photodynamic and laser therapy have led to further inves- Visible light (400–760 nm) is the wavelength range of general
tigations to be conducted on the cutaneous effect of visible illumination. Other regions of this spectrum include radio
light; yet, until recently, technologies in the incoherent, waves, microwaves, infrared radiation (heat), UV, X-rays and
nonlaser light sources have had great difficulty in fraction- gamma radiation.
ating light to produce pure visible spectrum without a UV
Definition of visible radiation

†This paper is part of a special issue dedicated to Professor Hasan Mukhtar on


The visible spectrum is the portion of electromagnetic radiation
the occasion of his 60th birthday. visible to the human eye, which responds to wavelengths from
*Corresponding author email: hlim1@hfhs.org (Henry W. Lim) 400 to 700 nm; some individuals can perceive wavelengths from
 2008 The Authors. Journal Compilation. The American Society of Photobiology 0031-8655/08

450
Photochemistry and Photobiology, 2008, 84 451

380 to 780 nm. A photopic eye, namely the eye that has been UVB. Mediators released under the effect of visible light and
exposed to a relatively high intensity light which permits action spectrum of erythema in the visible range are not yet
photochemical changes in the retina and constriction of the fully studied. A 1960 study (11) described the erythema caused
pupil, has its maximum sensitivity at 555 nm, the green region by UV radiation and visible light. It found that the UV
of the optical spectrum. The frequently used term, UV light, is erythema caused by 250 and 297 nm is the result of capillary
technically incorrect. The term ‘‘light’’ is best used for dilatation, and such wavelengths penetrate only very superfi-
wavelengths of radiation that results in a sensation of vision. cially into the upper dermis. On the other hand, as the long-
Hence, the preferred term is UV radiation (8). wave UV (366 nm) and visible light (405 and 436 nm)
penetrate much deeper into the dermis, it is suggested that
the erythema caused by these longer wavelengths radiation is
Skin optics
the result of dilatation of the vessels of the subpapillary plexus.
Once photons enter the skin one of two processes may occur.
The first process that takes place when UV and visible photons
Radical production
enter the skin is absorption, which initiates chemical changes in
the cells. When absorbed, the energy of the photon is Ascorbate is a nonenzymatic antioxidant, which contains
transferred to the chromophore and the photon no longer oxidative products including ascorbate free radical (12). A
exists; hence the depth of penetration of radiation is affected by 2006 study (13) of ex vivo irradiation of human skin with solar-
position and absorption spectrum of the chromophore in the simulated radiation, showed that the ascorbate radical signal
skin. The second process to occur is scattering, which is was directly proportional to the irradiance. Radical produc-
dependent on the wavelength of the photon, which affects the tion in the substratum corneum by UV and visible light
depth of penetration of radiation into the skin (8). Absorption components were approximately 67% and 33%, respectively.
spectrum is the probability of absorption of photons against This study indicated that visible light contribution to radical
the wavelength. A number of chromophores only absorb UVB production, which has obvious implications in the design of
and UVA, and others absorb throughout the UV and visible organic sunscreens and the prevention of free-radical, induces
wavebands. b-carotene has absorption maxima at 465 and damage.
490 nm in the visible spectrum, but also absorbs in the UV
range. Protoporphyrin IX has an absorption maximum at
Pigmentation
405 nm but absorbs UVA strongly; the other major absorption
bands include 410, 504, 538, 576 and 630 nm. Other endoge- Limited information is available regarding the role of visible
nous chromophores which absorb visible light include melanin, light on pigmentation. As a result, most information is
water, riboflavin, hemoglobin and bilirubin. In photodynamic extrapolated from UVA wavelengths upon pigmentation. Skin
therapy (PDT), the photosensitizing dyes absorb longer visible pigmentation induced by radiation occurs in three
wavelengths (>650 nm), while 5-aminolaevulinic acid (ALA) phases—immediate pigment darkening (IPD), persistent pig-
PDT results in endogenous protoporphyrin IX production ment darkening (PPD) and delayed tanning (DT) (14). IPD
which absorbs light in the aforementioned spectra. Exception- occurs as a response to low doses of UVA (1–5 J cm)2),
ally, melanin absorbs throughout the UVB, UVA and visible appears immediately after exposure, and fades within 20 min
wavebands (8). Figure 1 illustrates absorption spectra of some to 2 h (15). IPD results from oxidation and redistribution of
of the chromophores in the skin. pre-existing melanin (14). At higher UVA doses (>10 J cm)2),
PPD occurs and persists from 2 to 24 h (15). PPD also results
from oxidation and redistribution of pre-existing melanin (14).
Erythema
In contrast to IPD and PPD, DT, occurring days after
UVB can penetrate through the epidermis to the upper dermis. exposure, is associated with the synthesis of new melanin. Both
It exerts its biologic effects, in part, via mediators released by UVB and UVA are capable of inducing DT, However, DT
epidermal cells (9,10). Erythema and sunburn are mainly induced by UVA is preceded by IPD and PPD without
caused by UVB radiation, although UVA and visible light can noticeable redness, while that induced by UVB is always
also cause skin erythema, requiring much higher doses than preceded by erythema. UVB is more efficient than UVA in
inducing erythema and DT (16).
In 1983, Kollias and Baqer (17) conducted an in vivo study
on the changes in pigmentation induced by visible and near-
infrared light using a polychromatic light source of 390–
1700 nm, which simulates the solar spectrum without the UV
part, with powers of up to 0.35 W cm)2. Their aim was to
determine the changes in color occurring during irradiation and
to also record them using remittance spectroscopy. They found
that pigmentation could occur without any UV component,
and that there were no erythema or temperature changes
greater than 0.4C, even after 3 h of irradiation, associated with
the pigmentary change. They objectively defined detectable
erythema as changes in the remittance at 542 and 575 nm of
more than 5%. IPD was observed with energy greater than
Figure 1. Skin chromophore absorption spectra. 720 J cm)2, and the pigmentation lasted for up to 10 weeks.
452 Bassel H. Mahmoud et al.

Other studies showed that exposure of normal skin to 320 nm) on the skin are mediated predominantly by direct
visible light can result in the induction of IPD, immediate DNA damage, while the effects of UVA (320–400 nm) are
erythema and DT. Porges et al. (18) exposed 20 healthy dominated by indirect damage caused by reactive oxygen
individuals with skin Types II, III and IV to visible light source species (ROS) such as singlet oxygen. UVA has been divided
of a compact 150 W xenon-arc solar simulator, with a spectral into UVA1 (340–400 nm) and UVA2 (320–340 nm), with
distribution between 385 and 690 nm. Both IPD and imme- UVA2 inducing damages similar to that of UVB (20). UVB
diate erythema faded over a 24-h period; whether these dominates the carcinogenic effects of sunlight; however, UVA
represent thermal effects is unclear. The residual tanning has been estimated to contribute around 10–20% to the
response remained unchanged for the remaining 10-day carcinogenicity of sunlight (21).
observation period. The threshold dose for IPD with visible Similar to the continuum of the biologic effects of UVB to
light was between 40 and 80 J cm)2, while the threshold dose UVA2, because the division between UVA1 and visible light
for DT was closer to 80–120 J cm)2. is arbitrary, the photobiologic effects of UVA1 and visible
Because of lack of standardization pertaining to the light may also be similar (22). The effects of UVA1 and
spectrum of visible light producing sources in the aforemen- visible light on human skin have not been comprehensively
tioned studies by Kollias and Porges, it is hard to compare investigated, although it has been shown that UVA1 (365 nm)
their results. The filter used in the latter study was a 3-mm induces squamous cell carcinomas (SCC) in hairless mice
Schott GG385, which should remove most of the short which have been exposed to a daily radiation at three
wavelength UV radiation; however, a part of the long UVA different doses—240, 140 and 75 kJ m)2—for 620 days (23).
spectrum, from 385 to 400, together with the visible light, is Due to the lack of in-depth investigations regarding the long-
still emitted by this filtered light source. term effects of visible light on skin, and as it is widely used in
While currently there is no standardized visible light source laser and PDT, further studies on its long-term effects are
used consistently in all studies, there are several ways of needed.
obtaining visible light radiation. Broadband visible light could Edstrom et al. (22) studied the effects of repetitive low doses
be produced by filtering out the UV and infrared energy in of UVA1 on 12 healthy individuals, utilizing an output
most broadband light sources. For example, the solar simu- spectrum between 340 and 400 nm, a peak emission at
lator could be used with proper filters to attenuate the UVA, 365 nm, and visible light using an Osram xenon-arc lamp
UVB and IR radiation. Long-pass glass filters could be used to with two filters (Schott KG 3 and GG 420), which gives a
attenuate UVA and UVB radiation and heat-absorbing hot transmission of only visible light. A segment of the buttock
mirrors could be used to block the infrared energy. A low-cost was exposed to 20 J cm)2 UVA1 and another segment to
option is to use an incandescent lamp with a Plexiglas that 126 J cm)2 of visible light three times a week for 4 weeks.
blocks most of the UV component. The more expensive option Edstrom et al. observed a dispersed pattern of p53 (involved in
is to use a xenon-arc lamp with modern high-throughput several cell-cycle checkpoints, in G1, G2 and at mitosis)
interference filters (Semrock, Inc., Rochester, MN) to obtain positive cells in the epidermis, which may be a reversible
the broadband visible light (400–700 nm) while efficiently reaction to both UVA1 and visible light. Ren et al. (24)
blocking the UV radiation as much as 6 orders of magnitude. observed a compact pattern, which reflected a clonal multipli-
To minimize the effect of heat resulting from infrared cation of keratinocytes with mutated p53; no expression of
radiation, the ideal choice could be a water filter (Y. Liu, p21WAF)1 (the p53 mediator responsible for the arrest in G1)
personal communication). In the future, it would be ideal to in keratinocytes was noted, which is in contrast to that
use an identical type of light source in all studies. reported after UVA and UVB irradiation (25).
Kollias et al. (19) reported the development of erythema Interestingly, p53 was immunohistochemically detectable in
and melanogenesis in skin Type V individuals following the biopsy specimens of healthy individuals after only few
exposure to 295, 305, 315 and 365 nm monochromatic irradiations with UVA1 in suberythemal doses. The increase
radiation, and then compared erythema and melanogenesis was observed even after the first UVA1 exposure, and there
observed in individuals with skin Types I and II. The ratio of were more positive cells after each consecutive irradiation. The
values for the minimum erythema dose between skin Type V number of p53-positive cells tended to accumulate and
and skin Types I and II was 2.29 ± 0.8 (mean ± SD), which remained elevated 2 weeks after the last irradiation. An
is close to the ratio of constitutional pigment in these skin increased expression of cyclin A and a preferential expression
types, as measured by diffuse reflectance spectroscopy in the of Ki67 was also observed, indicating proliferation of epider-
visible range. Interestingly, the minimum melanogenic dose at mal cells, which is a known effect of UVA1. These findings
295, 315 and 365 nm was independent of skin type. suggest that there was an increase in epidermal cells in the G1
It would be interesting to correlate the above data presented phase. p53 downregulates the bcl-2 gene, whose product acts as
by Kollias and colleagues with future studies in the longer an antiapoptotic agent. However, these investigators noted a
visible light spectrum aiming at determining the minimal small increase in bcl-2 expression, the opposite of what was
erythema dose and minimum melanogenic dose at different expected, indicating that p53 may be mutated. Visible light
wavelengths. also caused an increase in p53-positive cells and proliferation
in the epidermis, but to a lesser degree than after irradiation
with UVA1 (22).
CELLULAR AND MOLECULAR
Kielbassa et al. (26) studied action spectrum for the
PHOTOBIOLOGY
formation of dimers and oxidative DNA modification in
Radiation has a variety of biologic effects on human skin, mammalian cells, using a monochromatic radiation. Oxidative
depending on the wavelength. The effects of UVB (290– DNA damage formation was observed extending from the
Photochemistry and Photobiology, 2008, 84 453

UVA1 range into visible light, with a peak between 400 and
Solar urticaria
450 nm.
Similar to long-wave UVA, visible light produces DNA Solar urticaria is a rare photosensitivity disorder, most
damage indirectly through the generation of ROS. Hoffmann- commonly occurring in early adulthood, but with variable
Dorr et al. (27) have analyzed the generation of micronuclei age of onset. The disease has been well described from the
associated with the induction of oxidative DNA damage by beginning of the 20th century. Hundreds of cases have been
visible light (>395 nm) in melanoma cells and primary reported worldwide. In 1988, Champion (30) reviewed 2310
culture of human skin fibroblasts. Using cyclobutane pyrim- cases of urticaria which his group had seen over a period of
idine dimers as a marker of the direct effect of electromag- 30 years, of which nine (0.4%) developed solar urticaria. In a
netic radiation, and the presence of modified DNA bases recent survey of 390 patients with urticaria, only two cases
sensitive to repair enzyme glycosylase as a marker of (0.5%) of solar urticaria were found. In a study conducted in a
electromagnetic radiation-induced oxidative damage, it was photodermatology referral center, the percentage of patients
concluded that indirectly generated oxidative DNA modifica- with solar urticaria in relation to other photodermatoses
tions can contribute significantly to the adverse effects ranged from 4% to 18% (31).
of visible light. Consequently, the wavelength dependence of Solar urticaria is considered to be a Type I immediate
DNA mutation induction does not correlate with that of hypersensitivity response, mediated by mast cells. It is char-
pyrimidine dimer formation. The ratio of pyrimidine dimers acterized by the onset of a pruritic, erythematous and
and oxidative DNA modifications at various wavelengths urticarial eruption on sun-exposed skin within minutes fol-
depends on both the direct and indirect mechanisms which lowing sun exposure, and is usually resolved within 2 h, if
have an impact on the assessment for the risk of mutation further exposure is avoided. Solar urticaria is usually more
related to solar irradiation. In addition to a peak at 313 nm, marked on skin that is normally covered, with relative sparing
DNA base modifications by solar radiation has a second peak of the face and dorsa of the hands. Episodes may be
between 400 and 450 nm because of the excitation of accompanied by systemic symptoms such as headache, nausea,
endogenous photosensitizers. Oxidative damage induced by bronchospasm, faintness and syncope (32).
400–500 nm accounts for approximately 10% of the total
endonuclease-sensitive base damage in cells exposed to Action spectrum of solar urticaria. The activating wavelengths
sunlight (26). responsible for solar urticaria vary from UVB to visible light.
In an in vivo study by Kvam and Tyrrell (28), the total Monochromator phototesting for solar urticaria was per-
amount of oxidation of guanine induced by monochromatic formed by Stratigos et al. (33) in 23 patients, the majority
radiation ranging from a UVB wavelength (312 nm) up to exhibiting induction of lesions with visible light. In a study by
wavelengths in the near-visible (434 nm) range in dermal Uetsu et al. (34), 24 patients (60%) were sensitive only to
fibroblasts was the same or greater than the amount of visible light. On the other hand, patients with solar urticaria
cyclobutane pyrimidine (the major type of direct DNA seen in the UK and Europe have been reported to be activated
damage). The action spectra for oxidative damage in skin most frequently by UV radiation. The majority (77%) of
fibroblasts were UVA (above 334 nm) and near-visible radi- patients reported by Frain-Bell (35) reacted to a wide action
ations. In rapidly dividing lymphoblastoid cells, no oxidative spectrum from UVB to visible light, and only 5 of 26 patients
guanine damage was induced. However, in melanoma cells, responded to visible light alone. Ryckaert and Roelandts (36)
almost as much damage as in nongrowing fibroblasts (1.1 per reported that from a series of 25 patients, 5 were sensitive only
104 guanine bases after 1200 kJ m)2 UVA) were found. The to visible light. Figure 2 displays the results of the aforemen-
authors concluded that oxidative DNA base damage can most tioned studies in relation to the action spectrum of solar
likely contribute to the induction of both nonmelanoma and urticaria.
melanoma skin cancer by sunlight. In solar urticaria, a certain spectrum outside of the
activating wavelengths may affect wheal formation. An
inhibition spectrum was first demonstrated by Hasei and
ROLE OF ELECTROMAGNETIC Ichihashi (37) in a patient who developed urticaria in the 400–
RADIATION IN THE PATHOGENESIS OF 500 nm range. Longer wavelengths inhibited wheal formation
PHOTODERMATOSES when irradiation was performed immediately after exposure to
UVB (290–320 nm) is the major spectrum responsible for radiation at the action spectrum. The wavelengths of the
cutaneous erythema, whereas UVA (320–400 nm) is involved inhibition spectrum are commonly longer than those of the
in the majority of photodermatoses. action spectrum. However, in one of their patients and also in
Photodermatoses are a broad group of skin disorders a patient studied by Leenutaphong et al. (38), the urticarial
primarily caused or exacerbated by exposure to UV or visible reaction to visible light was inhibited by pre-exposure and
light. Photodermatoses can be classified into five general postexposure to UVA, respectively. In addition, augmentation
categories—idiopathic, most likely immune mediated; second- spectrum has also been described in one patient with an action
ary to exogenous agents; secondary to endogenous agents; spectrum in the 320–420 nm range; exposure to longer
photoexacerbated dermatoses; and genodermatoses (29). wavelengths (450–500 nm) before exposure to 320–420 nm
Photodermatoses can have action spectrum in the UVB, enhanced wheal formation (34). In a study of 14 patients, an
UVA and ⁄ or visible light range, with UVA being the most augmentation spectrum was detected in four cases examined
common. Those with action spectrum in the visible light range, (29%), while an inhibition spectrum was found in 68%. The
solar urticaria, chronic actinic dermatitis (CAD) and cutane- mechanisms and clinical relevance of inhibition and augmen-
ous porphyrias will be discussed in the following section. tation spectra remain unknown (34).
454 Bassel H. Mahmoud et al.

Chronic actinic dermatitis


Chronic actinic dermatitis, a term originally proposed by
Hawk and Magnus (39), is a spectrum of clinical entity
covering conditions previously known as persistent light
reactivity, actinic reticuloid (40), photosensitive eczema (41)
and photosensitivity dermatitis (42). It affects most commonly
elderly males and manifests as a chronic eczematous photo-
distributed eruption (43). In several studies conducted in
photodermatology referral centers, the percentage of patients
with CAD in relation to other photodermatoses ranged from
5% to 17% (31).

Action spectrum of chronic actinic dermatitis. While the action


spectrum of CAD is primarily in the UVB or UVB and UVA
range, action spectrum at the visible light range has been
reported. Stratigos et al. (33) found that phototesting in
diagnosed cases of CAD showed abnormal erythemal reac-
tions to UVA (5 ⁄ 15, 33.3%), UVA and UVB (5 ⁄ 15, 33.3%),
UVB (2 ⁄ 15, 13.3%), and to UVA, UVB and visible light (2 ⁄ 15,
13.3%). Dawe and Ferguson (44) described a study involving
507 CAD patients investigated by monochromator phototest-
ing in the Dundee photobiology unit. UVB, UVA and visible
light induced lesions in 95%, 92% and 67% of cases,
respectively. Phototesting is always abnormal in moderate to
severe CAD, confirming the disorder. Papular or eczematous
lesions are commonly seen after exposure to the UVB
wavelengths, often also the UVA but rarely with visible light.
Occasional abnormalities have been reported to just the UVA,
and also very rarely to just 600 nm visible light (45).

Phototoxic and photoallergic skin reactions


Phototoxic reactions are significantly more common than
photoallergic reactions; they mimic exaggerated sunburn. Pho-
toallergic reactions are delayed hypersensitivity response
resembling allergic contact dermatitis on sun-exposed areas,
but may also extend into covered areas (46). Management
includes topical and systemic corticosteroids; avoidance of
precipitating agents is essential.

Porphyrias
Congenital erythropoietic porphyria was described using
different terminology such as pemphigus leprosus by Schultz
in 1874 (47) (who noticed the dark coloration of urine);
xeroderma pigmentosum by Gagey in 1896 (48); hydroa
vacciniforme by M’Call Anderson in 1898 (49) (who was the
first to recognize that the lesions were induced by light);
hereditary syphilis by Vollmer in 1903 (50); hydroa aestivale
by Ehrmann in 1905 (51) (who first suggested that the lesions
resulted from the sensitization of the skin to light exposure
by porphyrins); and also by Linser in 1906 (52); until
Günther described the condition, in 1911, as a porphyria. The
effect of porphyrins in leading to acute photosensitivity was
demonstrated by Meyer-Betz when, in 1912, he injected
himself with 200 mg hematoporphyrin and became acutely
photosensitive (53). The name porphyria cutanea tarda was
Figure 2. Studies showing action spectrum of solar urticaria: (a)
Stratigos et al. (33). (b) Uetsu et al. (34). (c) Frain-Bell (35). (d) first used in 1937 by Waldenstrom (54), who also extensively
Ryckaert and Roelandts (36). studied acute intermittent porphyria. The other porphyrias
were described later, some after World War II. In 1951,
Photochemistry and Photobiology, 2008, 84 455

erythropoietic protoporphyria was recognized. Variegate blister formation (which is the most commonly seen type of
porphyria was recognized in South Africa in the 1940s and cutaneous porphyrias) and accumulation of the lipophilic
1950s (55). protoporphyrin which is characterized by an immediate
Porphyrias are caused by accumulation of endogenous burning sensation in the skin to exposure of light. This can
phototoxic agents (porphyrins) because of enzyme defects in be followed by swelling, redness, purpura and sometimes
heme biosynthesis. Characteristic porphyrin profiles in plasma, erosions. These clinical features occur more commonly in
erythrocytes, urine and stool allow for diagnosis. Porphyrin erythropoietic protoporphyria (64).
molecules are tetrapyrrole ring structures which absorb visible
light, generating excited states. Exposure of porphyrins to
sunlight results in the generation of free radicals, which cause PHOTOPROTECTION
lipid peroxidation and protein cross-linking leading to cell
Sunscreens
membrane damage and death. The type of cellular damage
depends on the solubility and tissue distribution of porphyrins Sunscreens are the mainstay of treatment for photodermatoses.
(56). In some cases, patients are sensitive to visible as well as UV
Most types of porphyrias are inherited in an autosomal radiation or may be sensitive to isolated visible wavelengths,
dominant manner with incomplete penetrance, but autosomal particularly in the blue region. Unfortunately, currently avail-
recessive and more complex patterns of inheritance are also able sunscreens are not effective for photodermatoses that
seen in some. The genes coding for the enzymes have been have action spectrum in the visible light range. Patients
characterized and localized; mutations in them have been taking systemic photosensitive drugs such as Photofrin for
identified in patients with porphyria. Individual porphyrias are PDT have photosensitivity in the visible light range for up to
genetically heterogeneous but the clinical phenotype is uni- 6 weeks (65).
form; the seven different porphyrias can be divided into two UV filters are divided into organic (also known as
subgroups—hepatic or erythropoietic—according to the organ chemical) and inorganic (also known as physical) filters;
in which accumulation of porphyrins and their precursors currently there is no effective organic filter for visible light.
primarily appears (57). Porphyrias occur from the deficiencies Chemical agents such as dibenzoylmethanes and camphor
of seven enzymes in heme biosynthesis. Aminolevulinate derivatives have been used to provide extended protection
dehydratase deficiency porphyria and acute intermittent por- for the UVA region. The term ‘‘total sunblock’’ is imper-
phyria do not have cutaneous findings. Cutaneous findings in fectly used to describe them. Only optically opaque filters
the other porphyrias could be subdivided into acute photo- are able to absorb visible light; therefore, only optically
toxicity and subacute phototoxicity (58). Prenatal diagnosis is opaque inorganic filters can protect against visible light. The
possible for congenital erythropoietic porphyria (59), and in two generally available inorganic sunscreen agents are zinc
vitro gene therapy has been successfully performed for oxide (ZnO) and titanium dioxide (TiO2) (66). When an
hepatoerythropoietic porphyria, congenital erythropoietic por- incident visible light enters particles of TiO2 and ZnO, it is
phyria and erythropoietic protoporphyria (60). reflected by some facets of the particles in the direction of
our eyes and consequently it appears white (67). Absorption
Pathogenesis of skin symptoms in hepatic porphyrias. Porphyria range from visible to UVA to UVB depends on the particle
cutanea tarda, hereditary coproporphyria and variegate por- size. To enhance their cosmetic acceptability, inorganic
phyria have chronic manifestations usually days after sun sunscreen agents are more commonly used in ‘‘micronized’’
exposure. Blistering and scarring occur on the backs of the or ‘‘microfine’’ form, and coated with dimethicone or silica
hands. Hypertrichosis and hyperpigmentation arise on the to maintain their effectiveness as sunscreen. Micronization
face. Skin symptoms are marked in summer after sun results in particle size of less than 50 nm in diameter, which
exposure. Skin symptoms develop as a result of interaction makes them much less visible on the skin; however, as the
of solar radiation (around 400 nm) with high amounts of pigment is micronized, absorption range shifts toward UVB,
circulating porphyrins, which originate from the liver and especially with TiO2, which makes them poor absorbers of
accumulate in the skin (61). One of the properties of visible light. Micronized ZnO has better UVA1 protection
porphyrins is their intense spectral absorption at about and lower refractive index compared to microfine TiO2;
400 nm. This is known as the Soret band, which is a very therefore it looks less white (66). Micronized forms of metal
strong absorption band in the blue region of the optical oxides not only scatter and reflect light, but also absorb UV
absorption spectrum of porphyrin. It is intense and narrow as they are capable of mobilizing electrons within their
and is most well developed in acid solution; its position and atomic structure. They are stable, nontoxic and safe in their
magnitude differ for each porphyrin (62). A study by Stolik coated form (68).
et al., on the kinetic of porphyrins in mice using photoacoustic Nano-sized formulations are able to enhance or reduce skin
and fluorescence spectroscopies, showed that the highest penetration at a limited rate. Insoluble TiO2 or ZnO nano-
porphyrin concentration in skin, determined from the optical particles (NP) which are colorless do not penetrate into the
absorption of the Soret band at 410 nm, occurred 2 h after viable portion of human epidermis. In vitro cytotoxicity,
delta-aminolevulinic acid administration and corresponds to genotoxicity and photogenotoxicity studies on insoluble NP
porphyrin production in skin tissue. In addition, a first peak which report uptake by cells, oxidative cell damage, or
was observed at 15 min, which possibly represents the por- genotoxicity should be interpreted with caution as these
phyrin content in blood vessels within the skin (63). Skin toxicities may be secondary to phagocytosis of mammalian
manifestations of porphyrias include two types—accumulation cells exposed to high concentrations of insoluble particles.
of water-soluble uroporphyrins and coproporphyrins with Other NP may have characteristics enabling skin penetration.
456 Bassel H. Mahmoud et al.

There is little evidence that NP have greater effects on the skin fibroblasts. Pre-exposure incubation with this concentration
or produce toxicities relative to micro-sized materials. Current of EGCG also significantly reduced UVA ⁄ VIS-induced dam-
evidence suggests that nano materials such as nano-sized age in skin fibroblasts and epidermal keratinocytes. Data also
vesicles or TiO2 and ZnO NP (currently used in sunscreens) show that green tea polyphenols can exert a protective role
have no risk to human skin (69). through dietary supplementation (75).
In one study, diffuse spectral transmittance of various
thicknesses (7–260 lm) of opaque sunscreen formulations were
PHOTOTHERAPY USING VISIBLE LIGHT
measured in the 350–800 nm range using a spectrophotometer.
Transmission through 20% ZnO paste was high and decreased Intense pulsed light therapy
minimally despite large increases in the sunscreen layer
In 1976, Muhlbauer et al. (76) described the thermocoagula-
thickness (up to 260 lm). Adding another visible light
tion of capillary hemangiomas and port-wine stains by
absorber, iron oxide, substantially lowered transmittance
polychromatic infrared light. It was not until 1990 that
below that predicted by the product of the transmittances for
Goldman and Eckhouse developed new high-intensity flash-
each component alone. The addition of pigments greatly
lamps for treating vascular anomalies of the skin; and in 1994
enhances photoprotection and can produce a better match
the first intense pulsed light (IPL) technology was marketed.
with the patient’s skin color, which consequently increases
Theoretically, lasers are also a form of IPL; however, in
cosmetic acceptability (70).
clinical practice, the term IPL is commonly used to refer to a
Results of the study of Moseley et al. (71) confirmed the
high-intensity polychromatic incoherent light with a wave-
observation that pigmentary TiO2 plus ZnO exhibits relatively
length range from 515 to 1200 nm; by using different filters, a
uniform transmission in the UVB and UVA region, which
variety of wavelengths can be obtained with IPL devices (77).
extends protection into the visible region; this preparation has
The first law of photobiology, the Grotthus–Draper law,
been shown to provide protection for patients with sensitivity
states that light must be absorbed by tissue for a biologic effect
to the blue light region of the visible light.
to take place, whereas transmitted or reflected light has no
effect. The mechanism of action of light systems corresponds
Window glass to the selective photothermolysis that Anderson and Parrish
(78) described for the pulsed dye laser (PDL). The different
Standard glass filters out UVB, but transmits longer wave-
absorption maximums of the respective target structures allow
lengths radiation: UVA, visible light and infrared. Currently,
the right wavelengths to be selected for heating (>80C) and
filters are added for UVA and infrared radiation to provide
destruction to occur. Hemoglobin absorbs at a wavelength of
different levels of UV and infrared protection (72). Common
580 nm and melanin absorbs the entire visible spectral range
types of glass used in residential and commercial buildings are
(400–750 nm). The wavelength determines the absorption
summarized in Table 1.
behavior and the depth of penetration of light, which increases
with the wavelength. With different cutoff filters (515–
Antioxidants 755 nm), the optimal wavelength spectrum can be filtered
out to correspond to the depth of the target structure and to
Antioxidants are less potent than sunscreens in preventing
the patient’s skin type (77).
sunburn (73). The advantage of oral antioxidants is that they
protect the entire skin surface without being affected by
Advantages of IPL. The three main chromophores (hemoglo-
external factors such as washing, perspiration or rubbing.
bin, water and melanin) in human skin have broad absorption
Topical antioxidants diffuse poorly into the epidermis and are
peaks. Therefore laser monochromaticity is not a prerequisite
unstable (74). UVA ⁄ visible light (VIS) has been evaluated with
for selective heating (79). As a result, IPL can be a versatile
only ())-epigallocatechin-3-gallate (EGCG), which is the most
device to treat a variety of conditions in a cost-effective
effective chemopreventive component of green tea. Different
manner.
concentrations of EGCG were effective in preventing DNA
damage induced by UVR ⁄ VIS in cultured human lung
Disadvantages of IPL. Significant experience is needed when
working with IPL because of its wide range of wavelengths,
Table 1. Common types of glass used in residential and commercial
buildings. pulse durations and fluences. Because of lack of monochro-
maticity, the spectrum may not be consistent from pulse to
Transmission Transmission in pulse. The large handpieces and spot sizes used with IPL make
in the UV the visible light the device harder to maneuver than the laser, especially over
Glass types range (%)* range (%)†
irregular skin surfaces and discrete lesions (80). The decision to
Clear glass >72 >90 use an IPL or laser depends on the physician’s expertise with a
Tinted (heat-absorbing) glass 40 62 specific device and its availability.
Reflective glass 17 19
Low-emissivity glass 20 71
Laminated glass <1 79 Low-level light therapy
UV-blocking coated glass <1 80
Insulating glass <1 69 Low-level light therapy (LLLT) means to deliver doses lower
than the optimum value for a certain indication; this
Source: Modified from Tuchinda et al. (72). dose would produce a diminished therapeutic outcome. This
*Measured from 300 to 380 nm. †Measured from 400 to 780 nm. can be done using either coherent-light sources (lasers) or
Photochemistry and Photobiology, 2008, 84 457

noncoherent light sources (light-emitting diodes [LEDs]). is absorbed strongly by bilirubin (91). Different light sources
Whether the coherent monochromatic light of a laser is better exist for treatment of hyperbilirubinemia. Blue fluorescent
than noncoherent light in a defined wavelength range is still tubes, with an output in the blue spectrum (430–490 nm), are
controversial. LLLT is different from other types of laser by an effective light source for lowering serum bilirubin (92).
having a low intensity, which causes low temperature changes; LEDs provide high-intensity light in the blue portion of the
still, it can induce biologic effects, with minimal discomfort to visible spectrum that emit light through a narrow wavelength
the patient (81). band with a peak emission between 450 and 470 nm. They do
The mechanism of LLLT is absorption of red and near- not emit significant infrared or UV radiation and produce
infrared light by chromophores present in the protein compo- minimal radiant heat; the latter must be taken into account
nents of the respiratory chain located in mitochondria, mainly when administered to a naked infant (93).
cytochrome c oxidase. This is followed by subsequent photo-
dissociation of inhibitory nitric oxide from cytochrome c
Photodynamic therapy
oxidase (82) leading to increased enzyme activity (83),
increased electron transport (84) and increased production of While studying the influence of acridine orange on protozoa,
ATP (85). In addition, LLLT stimulates the expression of Oscar Raab, a medical student at the Department of Pharma-
genes related to cellular migration and proliferation; it also cology at the University of Munich, Germany, was the first to
alters the production of growth factors and cytokines (86). discover that the cell-killing effects of a drug were potentiated
The currently two main indications for LLLT are wound by the presence of light. In 1904, von Tappeiner used the term
healing and stimulation of hair growth. A study by Demidova- ‘‘photodynamic reaction’’ (94).
Rice et al. (81) suggested that LLLT stimulates mouse wound Photodynamic therapy is a therapeutic technique based on
healing by promoting contraction. They hypothesized that the the delivery of a photosensitizer followed by irradiation with
illumination delivered at 30 min postwounding induces fibro- visible light, as the light by itself is not sufficient to produce
blast–myofibroblast transition and, hence, wound contraction. significant photochemical reaction in the tissue. This reaction
They compared coherent monochromatic light from Helium ⁄ activates molecular oxygen to generate ROS, such as singlet
Neon laser (632.8 nm) and light from a broadband noncoher- oxygen, the hydroxyl radical, the superoxide anion and
ent light source (635±15 nm) using the same spot size and the hydrogen peroxide, which have a cytotoxic effect (95).
same fluence rate. They observed improved wound healing Depending on the amount and localization in the target tissue,
under both experimental conditions (He ⁄ Ne laser and ROS either modifies cellular functions or induces cell death by
635±15 nm lamp) compared to controls; but the difference necrosis or apoptosis (96). Most of the currently approved
between the light from different sources was not significant. clinical photosensitizers belong to the porphyrin family.
They also compared four different wavelength ranges of red Photosensitizers developed in the 1970s and early 1980s are
and near-infrared light centered at 635, 670, 720 and 820 nm. called first-generation photosensitizers (e.g. Photofrin). The
The most pronounced stimulation of wound healing was photosensitizers made since the late 1980s are called second-
obtained with 820 nm delivered from the same noncoherent generation photosensitizers such as ALA, which is the most
polychromatic light source. Regarding fluences, 2 J cm)2 had common photosensitizing agent used in dermatology, first
the largest positive effect, 1 and 10 J cm)2 improved healing to described by Kennedy and colleagues in 1990. New second-
a lesser extent while 50 J cm)2 had a negative effect on wound generation photosensitizers (e.g. benzoporphyrin derivatives,
healing. They concluded that the wavelength is the main phthalocyanines and chlorins) were then developed; they have
variable that defines the wound healing in response to light. the advantage of producing a photosensitivity that lasts for a
The other indication of LLLT is male and female pattern limited duration. Benzoporphyrin derivative has a delay before
hair loss. Bernstein (87) noticed thick terminal hair developed irradiation of up to 2.5 h and duration of photosensitization of
in the treated area following 810 nm diode laser treatment. He up to 7 days. Third-generation photosensitizers refer to the
concluded that this phenomenon should be studied to better modifications such as biologic conjugates (e.g. antibody
understand hair growth cycles and to help develop more conjugate, liposome conjugate) (97).
effective treatments for hair loss and hair growth. There are
still no controlled trials to assess the efficacy of LLLT for hair Photosensitizers. Topical sensitizers. Eosin red or erythrosine
growth, that is why its clinical use remains controversial. were the first topical photosensitizers used to treat conditions
such as pityriasis versicolor, psoriasis, molluscum contagio-
sum, syphilis, lupus vulgaris and skin cancer (94). ALA is not a
Phototherapy for hyperbilirubinemia
photosensitizer by itself but it is metabolized to photosensitiz-
Bilirubin alteration in the treatment of neonatal hyperbiliru- ing protoporphyrin IX (PpIX) through the intrinsic heme
binemia with phototherapy includes three different mecha- pathway. It penetrates through the stratum corneum when
nisms—photooxidation and photooxygenation to biliverdin, applied topically as a prodrug, and into dystrophic skin cells
maleimides and propentdyopents (88); photoaddition to pro- and the sebaceous gland where it is transformed into a highly
tein-bound bilirubin (89); and constitutional, configurational photoactive porphyrin derivative, PpIX (98). ALA synthesis
and structural photoisomerization. Production and excretion and accumulation is higher in malignant and premalignant
of structural photoisomers through bile and urine is considered cells than in their normal tissues. The ratio of PpIX accumu-
to be the main mechanism (90). lation in epidermal tumors in comparison with surrounding
Phototherapy lamps with output in the blue to green part of normal skin is 10:1 (99). The absorption spectrum of PpIX is
the spectrum are most effective in lowering serum bilirubin within the visible spectrum. Different light sources can be used
levels. At these wavelengths, light penetrates the skin well and to activate it, including those that emit blue (410 nm), yellow
458 Bassel H. Mahmoud et al.

(595 nm) or red (630 nm) light. PpIX has a high absorption sion of abnormal dermal capillaries; sebaceous hyperplasia, as
peak corresponding to the Soret band at about 405 nm and 5-ALA converts to PpIX in sebaceous glands; in photodamage
other absorption maxima, the Q-bands, at approximately 510, (dyspigmentation ⁄ poikiloderma), as ALA enhances the effec-
545, 580 and 630 nm. Although the Q-bands are 10- to 20-fold tiveness of laser and light treatment; and in actinic cheilitis
smaller than the peak in the Soret band, most clinical studies (109).
have used 625–633 nm red light, which allows for a deeper Fluorescence diagnosis. The selectivity of porphyrin follow-
penetration into the skin (100). ing topical application of ALA is used for diagnostic purposes
Systemic sensitizers. Systemic photosensitizers are delivered (through illumination of tissues with blue light at the Soret
intravenously as they do not penetrate well into the skin. band) to induce the emission of pink fluorescent light for
Hematoporphyrin and Photofrin were the first photosensitiz- delineation of the tumor because of high ratio of porphyrin
ers to be studied, which have absorption spectra with a small content in the tumor against the surrounding tissue (110). The
peak at 625–633 nm. Therefore, by using a red light source, dermatologist can then perform either a guided biopsy or a
they would produce a biologic effect at a depth of 5–10 mm. complete resection of the tumor, sparing healthy tissue (111).
These photosensitizers also have an absorption peak in the
far red (660–700 nm) or near-infrared (700–850 nm) regions
Laser emitting light in the visible range
where the depth of penetration into the tissues is up to
20 mm (100). In addition, near–infrared-absorbing photosen- The first laser was developed by Maiman (112) in 1959 using
sitizers should allow for the treatment of highly pigmented a ruby crystal to produce red light with a 694 nm
tumors, such as metastatic lesions of melanoma, which do wavelength. Cutaneous laser surgery was changed in the
not respond in the visible range, but not above 850 nm. 1980s with the introduction of the theory of selective
Although wavelengths above 850 nm have good depth of photothermolysis by Anderson and Parrish, which was used
penetration in the tissue, they are not commonly used for the destruction of a target in the skin with minimal
because their photon energy is too low to trigger a photo- thermal injury (78). Lasers emitting light in the visible range
chemical reaction (101). are shown in Table 2. Once laser energy is absorbed into the
Ideal photosensitizers. An ideal photosensitizer should be skin, three basic effects are possible—photothermal, photo-
available in the market with low toxicity without irradiation, mechanical or photochemical. Selective photothermolysis is
but powerful photocytotoxicity, selective toward target cells, an example of photothermal damage, in which light of a
long-wavelength absorbing, rapid removal from the body and wavelength (which is absorbed by a target chromophore)
easily administered through various routes (102). selectively destroys the chromophore if the fluence is high
Light sources. Light sources used for PDT belong to three enough and the pulse duration is less than or equal to the
groups—broadband lamps, diode lamps and lasers. Their thermal relaxation time (TRT). The TRT is the time taken
activity depends on the emission spectrum, irradiance, spatial for the target to dissipate half of the incident thermal
distribution and constancy of the output. Broadband lamps energy. Photoaccoustic effect is another means by which a
emit radiation in the visible and infrared ranges but with laser can destroy a chromophore, in which extremely rapid
insignificant UV emissions. LEDs have a high and reliable thermal expansion can lead to acoustic waves and subse-
emission in a narrow bandwidth of 20–50 nm without infrared quently photomechanical destruction of the absorbing tissue.
emission. Lasers produce high-energy monochromatic light of Laser-induced initiation of a chemical process is another
a specific wavelength. They provide for the exact selection of consequence of laser therapy. Photochemical effects serve as
wavelengths matching the absorption peak of the photosensi- the basis of PDT (113).
tizer and a highly homogeneous light beam but with a limited The main endogenous chromophores which absorb visible
treatment field (101). light include melanin, oxyhemoglobin and dexohemoglobin.
Applications of PDT in dermatology. Premalignant skin The three primary absorption peaks of oxyhemoglobin are
lesions such as actinic keratosis became the first approved within the visible range of the electromagnetic spectrum—418,
dermatologic indication of PDT, and nonmelanoma skin 542 and 577 nm. Vascular lesions are commonly treated with
cancers, including Bowen’s disease, are amenable to treatment 585 and 595 nm lasers which have the advantage of deeper
using ALA-PDT. The exception is SCC, especially the nonsu- dermal penetration without significant loss of absorption
perficial types, because of the high recurrence rates and
metastatic potential and lack of evidence to support its routine
treatment with ALA-PDT (103). A number of additional Table 2. Classification of lasers emitting light in the visible range
nonmalignant conditions (e.g. psoriasis, viral warts and hair based on their wavelengths.
removal) are currently under clinical investigation worldwide
Lasers emitting light in the visible range Wavelength (nm)
(104,105). Studies of ALA-PDT have also been conducted on
basaloid follicular hamartomas and cutaneous T-cell lym- Argon-pumped tunable dye 577 ⁄ 585 nm
phoma (106), also on both Mediterranean and HIV-related Copper vapor laser 511 (green) ⁄ 578 (yellow)
Kaposi’s sarcomas, as it is effective, can be repeated and is not fd Nd:YAG and KTP 532
associated with immunosuppression (107). PDT differs from PDL 585
Ruby 694
other cancer therapies in that the individual components of
Alexandrite 755
drug and light are inert unless they are combined (108). Other
common dermatologic conditions treated include acne vulga- Nd = neodymium; YAG = yttrium-aluminum-garnet; PDL =
ris, through accumulation of 5-ALA in the pilosebaceous unit, pulsed dye laser; fd = frequency doubled; KTP = potassium-titanyl-
particularly the sebaceous gland; rosacea, by selective occlu- phosphate.
Photochemistry and Photobiology, 2008, 84 459

(114). Light penetrates the dermis to a depth of 1.2 mm and biologic effects, visible light is used for the treatment of many
photocoagulates vessels of up to 100 lm in diameter. PDL has dermatosis and for cosmetic purposes through its integration
been reported as a successful treatment for viral warts, as it in phototherapy devices such as IPL, lasers and in PDT.
photocoagulates the dermal vessels and compromises the
viability of the human papillomavirus-infected epidermis Acknowledgement—Dr. Mahmoud has received a supporting grant
(115). PDLs with longer wavelengths (585, 590, 595 and from Johnson and Johnson, Skillman, NJ.
600 nm) and extended pulse durations (1.5–40 ms) have been
recently developed, which have deeper tissue penetration
capability using large spot sizes while retaining vascular REFERENCES
specificity (116). 1. Newton, I. (1672) New theory about light and colors. Philos.
The potassium-titanyl-phosphate (KTP) laser uses a neo- Trans. R. Soc. Lond. 80, 3075–3087.
dymium:yttrium-aluminum-garnet (Nd:YAG) crystal 2. Herschel, W. (1800) Investigation of the powers of the prismatic
colours to heat and illuminate objects. Philos. Trans. R. Soc.
(1064 nm) to produce light that is passed through a KTP
Lond. 90, 255–283.
crystal that frequency-doubles the wavelength to 532 nm. It 3. Ritter, J. (1801) Annalen für Physik. 7, 527.
has the advantage, over PDL, in producing minimal ery- 4. Willan, R. (1808) On Cutaneous Diseases. Johnson, London.
thema and no purpura in patients with facial telangiectasia 5. Rayer, P. (1835) Traite´ des maladies de la peau. Paris.
(117). Q switched (QS) lasers have a very low pulse duration 6. Home, E. (1820) On the black rete mucosum of the Negro, being
a defense against the scorching effect of the sun’s ray. Philos.
expressed in nanoseconds, and are used for benign epidermal Trans. R. Soc. Lond. 111, 1.
and dermal pigmented lesions and tattoos. Superficially 7. Davy, J. (1829) Observations on the effect of sun’s rays on the
located pigments are treated with shorter wavelength lasers human body. Trans Med-chir. Soc. Edin. 256.
(e.g. 510 nm PDL, 532 nm frequency-doubled QS Nd:YAG), 8. Diffey, B. L. and I. E. Kochevar (2007) Basic principles of pho-
tobiology. In Photodermatology (Edited by H. W. Lim, H.
while deeper pigments require longer wavelength (e.g. 694 nm
Hönigsmann and J. L. Hawk), pp. 15–27. Informa Healthcare
ruby, 755 nm alexandrite, 1064 nm QS Nd:YAG). The larger USA, Inc., New York.
the spot size used, the deeper the penetration of the laser 9. Everett, M. A., E. Yeargers, R. M. Sayre and R. L. Olson (1966)
(118). Penetration of epidermis by ultraviolet rays. Photochem. Photo-
The target for selective photothermolysis in laser epilation biol. 5, 533–542.
10. Young, A. R., C. S. Potten, C. A. Chadwick, G. M. Murphy, J.
is melanin in the hair follicle, with a thermal relaxation time L. Hawk and A. J. Cohen (1991) Photoprotection and 5-MOP
of 40–100 ms (113). Melanin within the epidermis represents photochemoprotection from UVR-induced DNA damage in
a competing site for follicular melanin for laser energy humans: The role of skin type. J. Invest. Dermatol. 97, 942–
absorption. Cooling of the skin with refrigerated gels, 948.
11. Rottier, P. B. and J. C. Van Der Leun (1960) Hyperaemia of the
cryogenic sprays or contact cooling devices built into the
deeper cutaneous vessels after irradiation of human skin with
laser hand piece decreases epidermal injury mainly in darker large doses of ultra-violet and visible light. Br. J. Dermatol. 72,
skin patients; it also allows delivery of higher fluences (119). 256–260.
Ruby (694 nm), alexandrite (755 nm), diode (800 nm) and 12. Loertzer, H., S. Bauer, W. Morke, P. Fornara and H. J. Bromme
Nd:YAG (1064 nm) lasers are all used for hair removal (113); (2006) Formation of ascorbate radicals as a measure of oxidative
stress: An in vitro electron spin resonance-study using 2,2-Azobis
the selection of the appropriate device depends on both the (2-amidinopropane) dihydrochloride as a radical generator.
color and thickness of the hair follicle as well as the patient’s Transplant. Proc. 38, 674–678.
skin type. 13. Haywood, R. (2006) Relevance of sunscreen application method,
With the swift development of laser technology and the visible light and sunlight intensity to free-radical protection: A
study of ex vivo human skin. Photochem. Photobiol. 82, 1123–
better understanding of light–tissue interaction, cutaneous
1131.
laser treatment indications will continue to expand over the 14. Rhodes, L. E. and H. W. Lim (2007) The acute effects of ultra-
coming years. violet radiation on the skin. In Photodermatology (Edited by H.
W. Lim, H. Hönigsmann and J. L. Hawk), pp. 75–89. Informa
Healthcare USA, Inc., New York.
CONCLUSION 15. Hönigsmann, H. (2002) Erythema and pigmentation. Photoder-
matol. Photoimmunol. Photomed. 18, 75–81.
The visible light part of the spectrum has not been thoroughly 16. Gange, R. W., A. D. Blackett, E. A. Matzinger, B. M. Sutherland
investigated for many years, compared to UV radiation, and I. E. Kochevar (1985) Comparative protection efficiency of
regarding both effects and side effects. However, data are UVA- and UVB-induced tans against erythema and formation of
enough that various biologic effects have been shown to be endonuclease-sensitive sites in DNA by UVB in human skin.
J. Invest. Dermatol. 85, 362–364.
exerted via visible light radiation including erythema, pigmen- 17. Kollias, N. and A. Baqer (1984) An experimental study of the
tation and radical production, as diverse endogenous and changes in pigmentation in human skin in vivo with visible and
exogenous chromophores have their absorption spectrum in near infrared light. Photochem. Photobiol. 39, 651–659.
the visible light range. It has also been shown that visible light 18. Porges, S. B., K. H. Kaidbey and G. L. Grove (1988) Quantifi-
cation of visible light-induced melanogenesis in human skin.
can induce indirect DNA damage through generation of ROS. Photodermatology 5, 197–200.
The ability to fractionate visible light is a recent phenomenon. 19. Kollias, N., Y. H. Malallah, H. al-Ajmi, A. Baqer, B. E. Johnson
As a result, advances have been made with respect to its and S. Gonzalez (1996) Erythema and melanogenesis action
biologic activity on the skin but still a great deal is unknown. A spectra in heavily pigmented individuals as compared to fair-
number of photodermatoses have an action spectrum in the skinned Caucasians. Photodermatol. Photoimmunol. Photomed.
12, 183–188.
visible light range, even though most of the currently available 20. Young, A. R. (1999) The molecular and genetic effects of ultra-
sunscreens offer, if any, weak protection against such relatively violet radiation exposure on skin. In Photodermatology (Edited
longer wavelengths. Fortunately, because of its cutaneous by J. L. Hawk), pp. 25–42. Arnold, London.
460 Bassel H. Mahmoud et al.

21. Kelfkens, G., F. R. de Gruijl and J. C. van der Leun (1990) Ozone 43. Roelandts, R. (1993) Chronic actinic dermatitis. J. Am. Acad.
depletion and increase in annual carcinogenic ultraviolet dose. Dermatol. 28, 240–249.
Photochem. Photobiol. 52, 819–823. 44. Dawe, R. S. and J. Ferguson (2003) Diagnosis and treatment of
22. Edstrom, D. W., A. Porwit and A. M. Ros (2001) Effects on chronic actinic dermatitis. Dermatol. Ther. 16, 45–51.
human skin of repetitive ultraviolet-A1 (UVA1) irradiation 45. Lim, H. W., W. L. Morison, R. Kamide, M. R. Buchness,
and visible light. Photodermatol. Photoimmunol. Photomed. 17, R. Harris and N. A. Soter (1994) Chronic actinic dermatitis. An
66–70. analysis of 51 patients evaluated in the United States and Japan.
23. de Laat, A., J. C. van der Leun and F. R. de Gruijl (1997) Car- Arch. Dermatol. 130, 1284–1289.
cinogenesis induced by UVA (365-nm) radiation: The dose-time 46. Lugovic, L., M. Situm, S. Ozanic-Bulic and I. Sjerobabski-Mas-
dependence of tumor formation in hairless mice. Carcinogenesis nec (2007) Phototoxic and photoallergic skin reactions. Coll.
18, 1013–1020. Antropol. 31(Suppl. 1), 63–67.
24. Ren, Z. P., F. Ponten, M. Nister and J. Ponten (1996) Two dis- 47. Schultz, J. H. (1874) Ein Fall von Pemphigus leprosus, kompliciert
tinct p53 immunohistochemical patterns in human squamous-cell durch Lepra visceralis. Inaugural dissertation, Greisswald.
skin cancer, precursors and normal epidermis. Int. J. Cancer 69, 48. Gagey, P. L. (1896) Cas d’hemoglobinurie au cours d’un xeroderma
174–179. pigmentosa. These de Paris.
25. Ponten, F., B. Berne, Z. P. Ren, M. Nister and J. Ponten (1995) 49. M’Call Anderson, T. (1898) Hydroa cacciniforme. Br. J. Der-
Ultraviolet light induces expression of p53 and p21 in human matol. 10, 1–4.
skin: Effect of sunscreen and constitutive p21 expression in skin 50. Vollmer, E. (1903) Uber Hereditare Syphilis und Hama-
appendages. J. Invest. Dermatol. 105, 402–406. toporphyrinuria. Archiv f Derm u Syph. 1xv, 221.
26. Kielbassa, C., L. Roza and B. Epe (1997) Wavelength dependence 51. Ehrmann, X. (1905) Hydroa aestivale. Archiv f Derm u Syph.
of oxidative DNA damage induced by UV and visible light. 1xxvii, 163.
Carcinogenesis 18, 811–816. 52. Linser, X. (1906) Hydroa aestivale and Hamatoporphyrinuria.
27. Hoffmann-Dorr, S., R. Greinert, B. Volkmer and B. Epe (2005) Archiv f Derm u Syph. 1xxix, 251.
Visible light (>395 nm) causes micronuclei formation in mam- 53. Cripps, D. J. (1986) Porphyria: Genetic and acquired. IARC Sci.
malian cells without generation of cyclobutane pyrimidine di- Publ. 77, 549–566.
mers. Mutat. Res. 572, 142–149. 54. Waldenstrom, J. (1937) Studien uber porphyrie. Acta Med.
28. Kvam, E. and R. M. Tyrrell (1997) Induction of oxidative DNA Scand. 82, 133.
base damage in human skin cells by UV and near visible radia- 55. Dean, G. (1971) The Porphyrias. Pitman Medical, London.
tion. Carcinogenesis 18, 2379–2384. 56. Poh-Fitzpatrick, M. B. (2000) Porphyrias: Photosensitivity and
29. Millard, T. P. and J. L. Hawk (2002) Photosensitivity disorders: phototherapy. Methods Enzymol. 319, 485–493.
Cause, effect and management. Am. J. Clin. Dermatol. 3, 239– 57. Moore, M. R., K. E. McColl, C. Rimington and A. Goldberg
246. (1987) Disorders of Porphyrin Metabolism. Plenum Publishing
30. Champion, R. H. (1988) Urticaria: Then and now. Br. J. Der- Corporation, New York.
matol. 119, 427–436. 58. Lim, H. W. and J. L. Cohen (1999) The cutaneous porphyrias.
31. Kerr, H. A. and H. W. Lim (2007) Photodermatoses in African Semin. Cutan. Med. Surg. 18, 285–292.
Americans: A retrospective analysis of 135 patients over a 7-year 59. Daikha-Dahmane, F., M. Dommergues, F. Narcy, M. C. Gubler,
period. J. Am. Acad. Dermatol. 57, 638–643. Y. Dumez, E. Gauthier, Y. Nordmann, C. Nessmann, G. Ter-
32. Harber, L. C. and D. R. Bickers (1989) Solar urticaria. In rasse and F. Muller (2001) Congenital erythropoietic porphyria:
Photosensitivity Diseases: Principles of Diagnosis and Treatment Prenatal diagnosis and autopsy findings in two sibling fetuses.
(Edited by L. C. Harber and D. R. Bickers), pp. 209–218. B. C. Pediatr. Dev. Pathol. 4, 180–184.
Decker, Inc., Toronto. 60. Richard, E., M. Mendez, F. Mazurier, C. Morel, P. Costet,
33. Stratigos, A. J., C. Antoniou, E. Papathanakou, M. Daboudi, P. Xia, A. Fontanellas, F. Geronimi, M. Cario-Andre, L. Taine,
K. Tranaka, K. Tsara and A. D. Katsambas (2003) Spectrum of C. Ged, P. Malik, H. de Verneuil and F. Moreau-Gaudry (2001)
idiopathic photodermatoses in a Mediterranean country. Int. J. Gene therapy of a mouse model of protoporphyria with a self-
Dermatol. 42, 449–454. inactivating erythroid-specific lentiviral vector without preselec-
34. Uetsu, N., H. Miyauchi-Hashimoto, H. Okamoto and T. Horio tion. Mol. Ther. 4, 331–338.
(2000) The clinical and photobiological characteristics of solar 61. Poh-Fitzpatrick, M. B. (1986) Molecular and cellular mechanisms
urticaria in 40 patients. Br. J. Dermatol. 142, 32–38. of porphyrin photosensitization. Photodermatology 3, 148–157.
35. Frain-Bell, W. (1985) Solar urticaria. In Cutaneous Photobiology 62. Jope, E. M. and R. O’Brien J (1945) Spectral absorption and
(Edited by W. Frain-Bell), pp. 51–55. Oxford University Press, fluorescence of coproporphyrin isomers I and III and the melting-
Oxford. points of their methyl esters. Biochem. J. 39, 239–244.
36. Ryckaert, S. and R. Roelandts (1998) Solar urticaria. A report of 63. Stolik, S., S. A. Tomas, E. Ramon-Gallegos and F. Sanchez
25 cases and difficulties in phototesting. Arch. Dermatol. 134, (2002) Kinetic study of delta-Ala induced porphyrins in mice
71–74. using photoacoustic and fluorescence spectroscopies. J. Photo-
37. Hasei, K. and M. Ichihashi (1982) Solar urticaria. Determina- chem. Photobiol. B, Biol. 68, 117–122.
tions of action and inhibition spectra. Arch. Dermatol. 118, 346– 64. Murphy, G. M. (1999) The cutaneous porphyrias: A review.
350. The British Photodermatology Group. Br. J. Dermatol. 140, 573–
38. Leenutaphong, V., E. Holzle, G. Plewig, B. Grabensee and 581.
B. Kutkuhn (1987) Plasmapheresis in solar urticaria. Photoder- 65. Dougherty, T. J., M. T. Cooper and T. S. Mang (1990) Cuta-
matology 4, 308–309. neous phototoxic occurrences in patients receiving Photofrin.
39. Hawk, J. L. and I. A. Magnus (1979) Chronic actinic dermati- Lasers Surg. Med. 10, 485–488.
tis—An idiopathic photosensitivity syndrome including actinic 66. Lim, H. and H. Honigsmann (2007) Photoprotection. In Photo-
reticuloid and photosensitive eczema. Br. J. Dermatol. 101(Suppl. dermatology (Edited by H. W. Lim, H. Hönigsmann and
17), 24.[proceedings] J. L. Hawk), pp. 267–278. Informa Healthcare USA, Inc., New
40. Ive, F. A., I. A. Magnus, R. P. Warin and E. W. Jones (1969) York.
‘‘Actinic reticuloid’’; a chronic dermatosis associated with severe 67. Kollias, N. (1999) The absorption properties of ‘‘physical’’ sun-
photosensitivity and the histological resemblance to lymphoma. screens. Arch. Dermatol. 135, 209–210.
Br. J. Dermatol. 81, 469–485. 68. Wolf, R., H. Matz, E. Orion and J. Lipozencic (2003) Sun-
41. Ramsay, C. A. and A. K. Black (1973) Photosensitive eczema. screens—The ultimate cosmetic. Acta Dermatovenerol. Croat. 11,
Trans. St. Johns Hosp. Dermatol. Soc. 59, 152–158. 158–162.
42. Frain-Bell, W., T. Lakshmipathi, J. Rogers and J. Willock (1974) 69. Nohynek, G. J., J. Lademann, C. Ribaud and M. S. Roberts
The syndrome of chronic photosensitivity dermatitis and actinic (2007) Grey goo on the skin? Nanotechnology, cosmetic and
reticuloid. Br. J. Dermatol. 91, 617–634. sunscreen safety. Crit. Rev. Toxicol. 37, 251–277.
Photochemistry and Photobiology, 2008, 84 461

70. Kaye, E. T., J. A. Levin, I. H. Blank, K. A. Arndt and R. R. 93. Maisels, M. J., E. A. Kring and J. DeRidder (2007) Randomized
Anderson (1991) Efficiency of opaque photoprotective agents in controlled trial of light-emitting diode phototherapy. J. Perinatol.
the visible light range. Arch. Dermatol. 127, 351–355. 27, 565–567.
71. Moseley, H., H. Cameron, T. MacLeod, C. Clark, R. Dawe and 94. Szeimies, R. M., J. Drager and C. Abels (2001) History of pho-
J. Ferguson (2001) New sunscreens confer improved protection todynamic therapy in dermatology. In Photodynamic Therapy and
for photosensitive patients in the blue light region. Br. J. Der- Fluorescence Diagnosis in Dermatology (Edited by P. G. Calzav-
matol. 145, 789–794. ara-Pinton, R. M. Szeimies and B. Ortel), pp. 3–16. Elsevier,
72. Tuchinda, C., S. Srivannaboon and H. W. Lim (2006) Photo- Amsterdam.
protection by window glass, automobile glass, and sunglasses. 95. Henderson, B. W. and T. J. Dougherty (1992) How does pho-
J. Am. Acad. Dermatol. 54, 845–854. todynamic therapy work? Photochem. Photobiol. 55, 145–157.
73. Hamanaka, H., Y. Miyachi and S. Imamura (1990) Photopro- 96. Szeimies, R. M., S. Karrer and C. Abels (2001) Photodynamic
tective effect of topically applied superoxide dismutase on sun- therapy in dermatology. In Dermatological Phototherapy and
burn reaction in comparison with sunscreen. J. Dermatol. 17, Photodiagnostic Methods (Edited by J. Krutmann, H. Honigs-
595–598. mann and C. Elmets), pp. 209–247. Springer, Berlin.
74. Kullavanijaya, P. and H. W. Lim (2005) Photoprotection. J. Am. 97. Moser, J. G. (1997) Definitions and general properties of 2nd &
Acad. Dermatol. 52, 937–958; quiz 959–962. 3rd generation photosensitizers. In Photodynamic Tumor Ther-
75. Morley, N., T. Clifford, L. Salter, S. Campbell, D. Gould and apy—2nd & 3rd Generation Photosensitizers (Edited by J. G.
A. Curnow (2005) The green tea polyphenol (-)-epigallocatechin Moser), pp. 3–8. Harwood Academic Publishers, London.
gallate and green tea can protect human cellular DNA from 98. Piacquadio, D. J., D. M. Chen, H. F. Farber, J. F. Fowler Jr, S.
ultraviolet and visible radiation-induced damage. Photodermatol. D. Glazer, J. J. Goodman, L. L. Hruza, E. W. Jeffes, M. R. Ling,
Photoimmunol. Photomed. 21, 15–22. T. J. Phillips, T. M. Rallis, R. K. Scher, C. R. Taylor and G. D.
76. Muhlbauer, W., G. Nath and A. Kreitmair (1976) Treatment of Weinstein (2004) Photodynamic therapy with aminolevulinic acid
capillary hemangiomas and nevi flammei with light. Langenbecks topical solution and visible blue light in the treatment of multiple
Arch. Chir. ??(Suppl.), 91–94. actinic keratoses of the face and scalp: Investigator-blinded,
77. Raulin, C., B. Greve and H. Grema (2003) IPL technology: phase 3, multicenter trials. Arch. Dermatol. 140, 41–46.
A review. Lasers Surg. Med. 32, 78–87. 99. Fritsch, C., B. Homey, W. Stahl, P. Lehmann, T. Ruzicka and H.
78. Anderson, R. R. and J. A. Parrish (1983) Selective phototherm- Sies (1998) Preferential relative porphyrin enrichment in solar
olysis: Precise microsurgery by selective absorption of pulsed keratoses upon topical application of delta-aminolevulinic acid
radiation. Science 220, 524–527. methylester. Photochem. Photobiol. 68, 218–221.
79. Anderson, R. R. (2003) Dermatologic history of the ruby laser: 100. Nyman, E. S. and P. H. Hynninen (2004) Research advances in
The long story of short pulses. Arch. Dermatol. 139, 70–74. the use of tetrapyrrolic photosensitizers for photodynamic ther-
80. Ross, E. V. (2006) Laser versus intense pulsed light: Competing apy. J. Photochem. Photobiol. B, Biol. 73, 1–28.
technologies in dermatology. Lasers Surg. Med. 38, 261–272. 101. Calzavara-Pinton, P. G., M. Venturini and R. Sala (2007) Pho-
81. Demidova-Rice, T. N., E. V. Salomatina, A. N. Yaroslavsky, I. todynamic therapy: Update 2006. Part 1: Photochemistry and
M. Herman and M. R. Hamblin (2007) Low-level light stimu- photobiology. J. Eur. Acad. Dermatol. Venereol. 21, 293–302.
lates excisional wound healing in mice. Lasers Surg. Med. 39, 102. Huang, Z. (2005) A review of progress in clinical photodynamic
706–715. therapy. Technol. Cancer Res. Treat. 4, 283–293.
82. Lane, N. (2006) Cell biology: Power games. Nature 443, 901–903. 103. Braathen, L. R., R. M. Szeimies, N. Basset-Seguin, R. Bisson-
83. Wong-Riley, M. T., H. L. Liang, J. T. Eells, B. Chance, M. M. nette, P. Foley, D. Pariser, R. Roelandts, A. M. Wennberg and C.
Henry, E. Buchmann, M. Kane and H. T. Whelan (2005) Pho- A. Morton (2007) Guidelines on the use of photodynamic therapy
tobiomodulation directly benefits primary neurons functionally for nonmelanoma skin cancer: An international consensus.
inactivated by toxins: Role of cytochrome c oxidase. J. Biol. International Society for Photodynamic Therapy in Dermatol-
Chem. 280, 4761–4771. ogy, 2005. J. Am. Acad. Dermatol. 56, 125–143.
84. Pastore, D., M. Greco, V. A. Petragallo and S. Passarella (1994) 104. Robinson, D. J., P. Collins, M. R. Stringer, D. I. Vernon, G. I.
Increase in <–H+ ⁄ e- ratio of the cytochrome c oxidase reaction Stables, S. B. Brown and R. A. Sheehan-Dare (1999) Improved
in mitochondria irradiated with helium-neon laser. Biochem. Mol. response of plaque psoriasis after multiple treatments with topical
Biol. Int. 34, 817–826. 5-aminolaevulinic acid photodynamic therapy. Acta Derm.
85. Karu, T., L. Pyatibrat and G. Kalendo (1995) Irradiation with Venereol. 79, 451–455.
He-Ne laser increases ATP level in cells cultivated in vitro. 105. Stender, I. M., R. Na, H. Fogh, C. Gluud and H. C. Wulf (2000)
J. Photochem. Photobiol. B, Biol. 27, 219–223. Photodynamic therapy with 5-aminolaevulinic acid or placebo for
86. Zhang, Y., S. Song, C. C. Fong, C. H. Tsang, Z. Yang and M. recalcitrant foot and hand warts: Randomised double-blind trial.
Yang (2003) cDNA microarray analysis of gene expression pro- Lancet 355, 963–966.
files in human fibroblast cells irradiated with red light. J. Invest. 106. Leman, J. A., D. C. Dick and C. A. Morton (2002) Topical 5-
Dermatol. 120, 849–857. ALA photodynamic therapy for the treatment of cutaneous T-cell
87. Bernstein, E. F. (2005) Hair growth induced by diode laser lymphoma. Clin. Exp. Dermatol. 27, 516–518.
treatment. Dermatol. Surg. 31, 584–586. 107. Calzavara-Pinton, P. G., R. M. Szeimies, B. Ortel and C. Zane
88. Lightner, D. A., W. P. Linnane 3rd and C. E. Ahlfors (1984) (1996) Photodynamic therapy with systemic administration of
Bilirubin photooxidation products in the urine of jaundiced photosensitizers in dermatology. J. Photochem. Photobiol. B,
neonates receiving phototherapy. Pediatr. Res. 18, 696–700. Biol. 36, 225–231.
89. Jori, G., E. Rossi and F. F. Rubaltelli (1980) Phototherapy-in- 108. Jori, G. (1996) Tumour photosensitizers: Approaches to enhance
duced covalent binding of bilirubin to serum albumin. Pediatr. the selectivity and efficiency of photodynamic therapy. J. Pho-
Res. 14, 1363–1366. tochem. Photobiol. B, Biol. 36, 87–93.
90. Onishi, S., K. Isobe, S. Itoh, M. Manabe, K. Sasaki, R. Fukuzaki 109. Alster, T. S. and S. S. Surin-Lord (2006) Photodynamic
and T. Yamakawa (1986) Metabolism of bilirubin and its therapy: Practical cosmetic applications. J. Drugs Dermatol. 5,
photoisomers in newborn infants during phototherapy. 764–768.
J. Biochem. (Tokyo) 100, 789–795. 110. Ericson, M. B., C. Sandberg, F. Gudmundson, A. Rosen, O.
91. Ennever, J. F. (1990) Blue light, green light, white light, more Larko and A. M. Wennberg (2003) Fluorescence contrast and
light: Treatment of neonatal jaundice. Clin. Perinatol. 17, 467– threshold limit: Implications for photodynamic diagnosis of
481. basal cell carcinoma. J. Photochem. Photobiol. B, Biol. 69, 121–
92. American Academy of Pediatrics Subcommittee on Hyperbilir- 127.
ubinemia (2004) Management of hyperbilirubinemia in the new- 111. Baumler, W., C. Abels and R. M. Szeimies (2003) Fluorescence
born infant 35 or more weeks of gestation. Pediatrics 114, 297– diagnosis and photodynamic therapy in dermatology. Med. Laser
316. Appl. 18, 47–56.
462 Bassel H. Mahmoud et al.

112. Maiman, T. (1960) Stimulated optical radiation in ruby. Nature vessels probed in vivo: The need for 1-10-millisecond laser pulse
187, 493–494. treatment. J. Invest. Dermatol. 105, 709–714.
113. Acland, K. M. and R. J. Barlow (2000) Lasers for the derma- 117. West, T. B. and T. S. Alster (1998) Comparison of the long-pulse
tologist. Br. J. Dermatol. 143, 244–255. dye (590-595 nm) and KTP (532 nm) lasers in the treatment of
114. Tan, O. T., S. Murray and A. K. Kurban (1989) Action spectrum facial and leg telangiectasias. Dermatol. Surg. 24, 221–226.
of vascular specific injury using pulsed irradiation. J. Invest. 118. Tanzi, E. L., J. R. Lupton and T. S. Alster (2003) Lasers in
Dermatol. 92, 868–871. dermatology: Four decades of progress. J. Am. Acad. Dermatol.
115. Tan, O. T., R. M. Hurwitz and T. J. Stafford (1993) Pulsed dye 49, 1–31; quiz 31–34.
laser treatment of recalcitrant verrucae: A preliminary report. 119. Altshuler, G. B., H. H. Zenzie, A. V. Erofeev, M. Z. Smirnov,
Lasers Surg. Med. 13, 127–137. R. R. Anderson and C. Dierickx (1999) Contact cooling of the
116. Dierickx, C. C., J. M. Casparian, V. Venugopalan, W. A. Farinelli skin. Phys. Med. Biol. 44, 1003–1023.
and R. R. Anderson (1995) Thermal relaxation of port-wine stain

Anda mungkin juga menyukai