You are on page 1of 17

Journal of Photochemistry and Photobiology C: Photochemistry Reviews 47 (2021) 100403

Contents lists available at ScienceDirect

Journal of Photochemistry and Photobiology C:


Photochemistry Reviews
journal homepage: www.elsevier.com/locate/jphotochemrev

How does the skin sense sun light? An integrative view of light
sensing molecules
Leonardo Vinicius Monteiro de Assis a,1,3 , Paulo Newton Tonolli b,1 ,
Maria Nathalia Moraes c , Maurício S. Baptista d,∗,2 , Ana Maria de Lauro Castrucci a,e,2
a
Department of Physiology, Institute of Biosciences, University of São Paulo, Brazil
b
Human Genome and Stem Cell Research Center, Institute of Biosciences, University of Sao Paulo, Brazil
c
Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Brazil
d
Department of Biochemistry, Institute of Chemistry, University of São Paulo, Brazil
e
Department of Biology, University of Virginia, Brazil

a r t i c l e i n f o a b s t r a c t

Article history: The consensus on the effects of excessive sun exposure on human health has long emphasized the negative
Received 3 October 2020 effects of solar UV radiation. Nevertheless, although UV radiation has been demonized, less is known about
Received in revised form 2 February 2021 the consequences of sun exposure while using sunscreen, which can lead to high visible light exposure. UV
Accepted 18 February 2021
and visible light play key roles in vitamin D synthesis, reduction of blood pressure, among other beneficial
Available online 22 February 2021
effects. In this review, we aim to provide a comprehensive view of the wide range of responses of the
human skin to sunlight by revisiting data on the beneficial and harmful effects of UV and visible light.
Keywords:
We start by exploring the interaction of photons in the skin at several levels including physical (depth of
Skin biology
UV radiation
photon penetration), chemical (light absorption and subsequent photochemical events), and biological
Visible light (how cells and tissues respond). Skin responses to sun exposure can only be comprehensively understood
Chromophores through a consideration of the light-absorbing molecules present in the skin, especially the light-sensing
Opsins proteins called opsins. Indeed, many of the cellular responses to sun exposure are modulated by opsins,
Photosensors which act as the “eyes of the skin”.
© 2021 Elsevier B.V. All rights reserved.

Contents

1. Sunlight: friend or foe? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2


2. Overview of skin biology and exposome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3. Upon absorption light photons generate excited states . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
4. How visible light and UV radiation interact with opsins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
4.1. Discovery of the opsins in the skin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
4.2. Photophysical properties of opsins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
4.3. Advancement in the field: a historical view of opsin functions in the skin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
4.3.1. UVR sensor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
4.3.2. Visible light sensor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
5. Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13

∗ Corresponding author at: Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Avenida Prof. Lineu Prestes 748, São Paulo, SP, 05508-000, Brazil.
E-mail address: baptista@iq.usp.br (M.S. Baptista).
1
Both authors had same contribution to this study and share the 1st authorship.
2
Both authors had same contribution to this study and share the senior authorship.
3
Current address: Institute of Neurobiology, Center for Brain, Behavior, and Metabolism, University of Lübeck, Germany.

https://doi.org/10.1016/j.jphotochemrev.2021.100403
1389-5567/© 2021 Elsevier B.V. All rights reserved.
L.V.M. de Assis, P.N. Tonolli, M.N. Moraes et al. Journal of Photochemistry and Photobiology C: Photochemistry Reviews 47 (2021) 100403

1. Sunlight: friend or foe?


Leonardo Monteiro de Assis holds a Bpharm (Federal
University of Ouro Preto, 2014) and a PhD in Physiol-
ogy (University of São Paulo, USP, 2019). During his PhD Unquestionably, spending excessive periods of time in the
and postdoctoral experience, Leonardo and his research sun is harmful to the skin and overall health. Current photo-
group in a pioneering fashion characterized the pho-
toreception (opsins) and temporal controlling systems protection strategies mainly focus on avoiding exposure to the
(clock genes) in normal and malignant melanocytes. Cur- ultra-violet portion of the sun spectra (UV, 200–400 nm). However,
rently, Leonardo is a research associate at the University the overwhelming focus on the harm of UV radiation neglects three
of Lübeck, Germany.
important aspects of sun-skin interactions: i. While UV has been
proven to be carcinogenic [1], there is no scientific evidence indi-
cating that only UV is dangerous to the skin; ii. A solid body of
evidence has highlighted the danger of excessive exposure to non-
UV wavelengths of light [2]; iii. Sunbathing also has many beneficial
Paulo Newton Tonolli is a biologist graduated from the effects [2,3].
Federal University of Sao Carlos (Brazil) and currently is a The American Academy of Dermatology has encouraged peo-
researcher scientist in the Human Genome and Stem Cell
Research Center at University of Sao Paulo (USP), Brazil.
ple to use UV protection on a daily basis, with a zero-tolerance
He received his Ph.D. degree in Sciences (Biochemistry) approach to sun exposure [1,3,4]. Many professional societies have
from the Institute of Chemistry at USP, investigating the endorsed similar views, promoting campaigns that are consistently
effects of visible light on the skin keratinocytes, focus-
centered on the negative effects of sunlight, though their pre-
ing on the photosensitization of endogenous molecules
and organelles and photodamage. His Master degree was scriptions vary in the recommended level of sun avoidance. These
studying mitochondrial DNA (mtDNA) repair and the role campaigns have prompted unrestricted sunscreen usage, disre-
of transcription factor A in the defense of mtDNA against
garding the many good effects of sun exposure on human health
oxidative stress.
in the right amounts.
In addition, users are often unaware that the sunscreens do
not block all wavelengths of sunlight, including those marketed as
Maria Nathalia Moraes has a major in Biological Sci-
ences by CES/JF, M.Sc. and PhD. in Physiology by the
offering “broad-spectrum protection” [5,6]. These sunscreens actu-
University of Sao Paulo. She worked in Dr. Castrucci’s ally only offer meaningful protection against UVB (280–320 nm)
lab for 12 years, where she studied the circadian clock and UVA (320–400 nm) radiation, which only represent around 2%
and opsins in non-mammalian and mammalian verte-
of the solar irradiance that reaches the Earth’s surface, leading to
brate models. She elucidated how light and temperature
is translated as a synchronizer factor in peripheral tissues a false sense of protection. It is not surprising that epidemiological
and which triggered signaling pathways ultimately lead to studies have reported a worldwide increase in the incidence of skin
clock gene modulation. During her post-doctoral position, cancer [1,4], despite the rising use of sunscreen [5]. While a num-
she demonstrated for the first time, a link between TRP
channels and clock genes in the modulation of mammalian ber of factors are behind this trend, the usual explanation tends
peripheral tissues related with energy metabolism. Cur- to be superficial, with claims that people do not correctly apply
rently, she holds a young research investigator position in sunscreen on the skin, frequently spreading just 25–30 % of the
Dr. Cipolla-Neto’s lab, focusing her investigation on the effects of chronodisruption
on the central nervous system and metabolic tissues, using animals that sponta- recommended sunscreen amount [3]. However, many researchers
neously develop glaucoma. Her main question is related to what are the short and have concluded that the current photoprotection guidelines are
long-term consequences of the lack of light exposure on health. ineffective because they lack a scientific basis and ignore a large
body of evidence reported in literature [2,6–8].
Mauricio S. Baptista PhD is professor of Biochem- The redness in the skin caused by sun exposure, also called ery-
istry at the University of São Paulo (USP, Brazil). He
thema or sunburn, arises mainly from the effect of UVB radiation,
earned Bachelor (1990) and Master (1992) degrees in
Biochemistry from USP and holds a doctoral degree although UVA and visible light also cause skin redness with lesser
(1996) in Chemistry from Marquette University (USA). intensity [2]. Skin erythema is an acute inflammatory response to
He did his post-doctoral training at UW-Madison (1997)
damage which is concentrated in the nuclear DNA of skin cells.
and was visiting professor (2006) at the Université
Joseph Fourier (Grenoble-France). His main interests The redness functions as a warning sign to get out of the sun. All
are photochemistry/photobiology, membranes/interfaces people have this response, though it is smaller and more difficult
and mechanisms of cell death, where he published over to measure in people with darker skin tones [9]. Sunscreens avoid
180 papers. He serves as co-editor in chief of J. Photochem
Photobiol. B and J.Photochem.Photobiol. (Elsevier) and is part this response by blocking UVB rays. Even so, using sunscreen does
of the board of Photochemical & Photobiological Sciences not prevent a darkening of the skin, which many find aesthetically
(RSC) and of Scientific Reports (Nature). He is a member of the The Academy of pleasing. This, too, is a natural protective mechanism to defend
Sciences of the State of São Paulo.
the skin against excessive sun exposure [10]. Current sunscreen
technology does not provide any protection against ∼98 % of the
Ana Maria de Lauro Castrucci graduated in Biology and
got her Master and PhD degrees in Physiology at the Uni-
solar irradiance, which is comprised of visible light and infrared
versity of Sao Paulo. Her post doctorate was made under radiation (IR). Photons in the visible and IR ranges interact with
the supervision of Prof Mac Eugene Hadley, at the Uni- the skin, culminating in a burst of free radicals in the skin [11].
versity of Arizona, during the 80’s. In 1999, she took a
Remarkably, visible light and a portion of IR called infrared A (IRA,
sabbatical leave of one year as a visiting scholar at the
Uniformed Services University of the Health Sciences, 760–1440 nm) radiation penetrate much deeper in the skin than
Maryland. Since 2005 she has collaborated with the Uni- UV rays (Fig. 1, Table 1). Thus, individuals using UV protection will
versity of Virginia. She retired in 2001 from the University
nevertheless suffer from the action of visible light and infrared radi-
of São Paulo as Full Professor of Physiology, but kept her
laboratory as Senior Professor, since then. She is a member ation, which generate reactive oxidants and can saturate the skin’s
of the Brazilian Academy of Sciences and of The Academy redox defenses [2,11–13]. Within the skin, melanocytes sense blue
of Sciences of the State of São Paulo and received the light and UVA radiation through opsins (Fig. 1) and trigger melano-
National Order of Scientific Merit awarded by the President of Brazil. She presently
holds research grants from the São Paulo Research Foundation, FAPESP, focusing the genesis [14,15]. A number of other questions which have not yet
following subjects: clock genes, opsins, light, temperature, endocrines and signaling been properly answered will be also noted below.
mechanisms in crustaceans, fish, amphibians, birds and mammals. Another misconception is that darker-skinned people are fully
protected from the sun. Melanin is a natural pigment that does
defend the skin against DNA damage, redness and carcinogenesis

2
L.V.M. de Assis, P.N. Tonolli, M.N. Moraes et al. Journal of Photochemistry and Photobiology C: Photochemistry Reviews 47 (2021) 100403

Fig. 1. A) Solar radiation and its different wavelengths exert several effects on the skin. UV radiation is subdivided into UVC, which is fully blocked by the atmosphere, UVB,
and UVA radiation. Upon contact with the skin tissue, light photons can be reflected or diffusely transmitted at the surface. As light photons penetrate deeper into the skin,
the following outcomes may take place: 1) photons may be diffusely reflected; 2) photons may penetrate the skin layers (directly transmitted light), which is dependent
on the wavelength (see panel B for better visualization); 3) photons may be dissipated as heat energy. B) Skin layers, cell types and appendices are represented. Opsins are
expressed in keratinocytes, melanocytes, and fibroblasts. Chromophores are found in the epidermis and dermis. The depth of UV radiation and light penetration in the skin is
shown (please refer to Table 1 and Fig. 3). Opsins and chromophores represent two venues for UV and light photons interaction, which may lead to physiological responses.
The reader is referred to the text and to the Graphical Abstract for more details on each biological process mediated by these photosensors. Figure created by the authors.

Table 1 reaching the skin per second by square centimeter of an exposed


Penetration of particular wavelength ranges of light into the skin. Infrared B (IRB,
area. Around 98 % (UV) of the photons reaching the skin are filtered
1440 – 3000 nm) and C (IRC, 3000 nm – 10000 nm) only interact with the first lay-
ers of the epidermis, thus showing a poor penetration capacity. Data values were or reflected by the sunscreen, while the remaining of sunlight pho-
approximate due to different techniques and methodologies and were based on tons will interact with the skin without any protection barrier [22]
[62,65,66]. (Fig. 1). This person will not show signs of sunburn and may even
Wavelength (nm) Tissue Penetration (mm)
get a “beautiful tan”, but he/she is not warned that the usual tanning
is triggered by damage in the skin.
300 < 0.5
The sun protection factor (SPF) is not a precise parameter to
350 0.8
400 1 ensure comprehensive skin protection from sunlight. The light
450 1.5 sources used to calculate SPF and to evaluate UVA protection only
500 2.5 expose people to UV. The design of these tests is therefore not
550 3
indicative of real-world conditions and may give consumers a false
600 4
650 4.5 sense of protection, since these tests do not measure the full effect
700 5.2 of solar radiation. Additionally, SPF calculations do not consider
750 5.5 the photoinduced generation of reactive oxidants and the resulting
Near Infrared (750–1400) > 5.5 oxidative stress [22]. Indeed, antioxidant compounds (e.g., vitamins
C and E) have been studied by the Baptista group and others, and
the inclusion of these compounds in sunscreen formulations has
from UVB radiation [16]. However, as demonstrated by Baptista’s improved photoprotection [22–24]. In any case, it is undeniable
group, melanin acts as a double-edged sword, since it also induces that a person sunbathing on the beach and correctly using com-
oxidative stress and indirect DNA lesions such as the premuta- mercially available sunscreen will not be protected against 98 % of
genic 8-oxo-dG when skin and hair are exposed to visible light and the photons that will come into contact with their skin [25] (Fig. 1).
UVA [17–19]. Thus, even though individuals with more melanin Even though such a sunbather will not show signs of sunburn and
can spend more time in the sun without noticeable acute effects may even get a beautiful tan, they will likely be unaware of the skin
in the skin, they are not immune from the chronic effects of sun damage caused by their tanning. Could ineffective photoprotection,
exposure. While their thicker dermis makes wrinkles less notice- then, be the reason for increasing levels of skin cancer cases? Many
able, people with dark skin are more vulnerable to depigmentation scientists are working to shed light on this question [6,8,18,26–28].
[20]. Even so, a much greater problem for Asian and African people Furthermore, currently available sunscreens have been shown
in temperate climates is increased rates of cardio-metabolic dis- to suffer from several drawbacks. Many of them contain organic
eases that appear to be partly due to inadequate sunlight exposure molecules that have been shown to be toxic to skin, such as oxy-
and, consequently, low levels of vitamin D production [21] (see benzone [29]. The Food and Drug Administration (FDA) in the
additional information below). Therefore, darker-skinned people US has requested more data on the safety of twelve compounds
should also be encouraged to get some unprotected sun exposure, found in sunscreen formulations: ensulizole, octisalate, homos-
though always avoiding excesses. alate, octocrylene, octinoxate, oxybenzone, avobenzone, cinoxate,
Now, imagine someone sunbathing on the beach and using dioxybenzone, meradimate, padimate O, and sulisobenzone [30].
correctly the present marketed sunscreens. During this time, this The negative effects of sunscreens may also include widespread
individual will receive billions and billions of photons from sunlight damage to the environment. It is estimated that 14,000 tons of sun-

3
L.V.M. de Assis, P.N. Tonolli, M.N. Moraes et al. Journal of Photochemistry and Photobiology C: Photochemistry Reviews 47 (2021) 100403

screens enter the oceans annually, from beachgoers and domestic describe and discuss the interaction of opsin receptors with UV and
sewage effluent [31]. Recent studies have shown that organic com- visible light.
pounds in sunscreens may be involved with bleaching and death Our goal in this review is to provide a global analysis of how
of coral reefs [31,32]. States like Hawaii (SB 2571, Act 104, March the skin senses light of different wavelengths through interactions
3rd, 2018) with its famous barrier reefs have prohibited the use of with chromophores and light- sensing molecules named opsins.
sunscreen with formulations that include oxybenzone [33]. We also examine the way in which the skin influences peripheral
Discussions of skin protections often neglect the benefits of sun organs after receiving light stimuli. Crucially, our approach deals
exposure for human health. Approximately 90 % of all vitamin D with the skin as an important part of a complex system, with a sig-
used in our body is formed through the action of UVB rays on nificant role due to its photosensitivity. We aim to provide a global
the skin, where vitamin D3 synthesis takes place [34]. Vitamin D view of how the skin senses light and UV radiation. Notably, we
plays a substantial function in regulating calcium and phospho- seek to determine whether chromophores or opsins are the pre-
rus levels, improving bone and muscle health, strengthening the dominantly responsible for skin photosensitivity, which has been
immune system, and preventing several types of cancer, cardio- a leading investigation by scientists in this area. We further call for
vascular diseases, depression and other illnesses [35–38]. While a reevaluation of skin’s sensory ability and its impact in skin phys-
estimates vary on the optimal time to spend in the sun, popula- iopathology. Finally, we highlight important gaps in the literature
tion studies show that people with fair skin, in temperate regions, and suggest future directions in the skin photobiology field.
should spend around 10−20 min sunbathing three times per week
with their full body exposed [38]. In contrast, the trend towards
spending one’s time largely indoors and not exposing oneself to 2. Overview of skin biology and exposome
sunlight without protection has resulted in an epidemic of vita-
min D deficiency [34–40]. In the US, the treatment of diseases The skin is the human body’s largest organ and the first
caused by vitamin D deficiency costs around US$ 16–25 billion, defensive barrier. In addition to its protective function, skin is
while the cost of skin cancer treatments amounts to US$ 8 billion sensitive to touch, pain, pressure, itching, temperature, and solar
[41,42]. radiation [55]. The skin is comprised of two layers, the epidermis
Largely missing in the discourse around sun exposure is a and dermis, which contain a number of cell types. The epidermis is
discussion of its beneficial effects beyond vitamin D synthesis. largely comprised of keratinocytes, which originate in epidermal
Understanding these effects is perhaps the most important con- stem cells. These cells migrate upwards and differentiate into the
tribution of this review. Skin cells such as keratinocytes and various layers of the outer skin: from the base to surface, these
melanocytes can detect the presence of sunlight using a set of pro- are the stratum basale, stratum spinosum, stratum granulosum,
teins called opsins, which are responsible for capturing light in stratum lucidum, and stratum corneum. As they migrate upwards,
photoreceptive organs such as the eyes [43] (Fig. 1). These proteins keratinocytes start to express different markers and proteins that
will be discussed further elsewhere in this review, but for now it is ultimately generate the epidermal barrier [55]. Melanin-producing
enough to mention that it is intriguing that sunlight photoreception cells named melanocytes reside in the stratum basale. Melanin
by skin triggers various physiological responses, such as melanin synthesis is stimulated in response to interaction with light and
production [14,15,44]. Indeed, it even regulates the cell activity of the pigment is transferred to neighboring keratinocytes, where
the immune system [45]. The body’s central biological clock, the it acts as a shield against UV radiation and a scavenger of free
suprachiasmatic nucleus located in the hypothalamus, adjusts itself radicals [56–58]. The dermis provides support and nourishment
each day using sun light [46,47], while disruptions to this rhythm to the epidermis and is mainly comprised of fibroblasts, lymph
can have negative health effects [47,48]. Low sun exposure can also and blood vessels, nerve endings, hair follicles, and glands. This
exacerbate conditions such as obesity and cardiovascular disease layer is subdivided into two sections: the papillary layer, which
[49]. Another important benefit of sun exposure is related to blood has nerve endings and capillaries, and the reticular layer, which
pressure levels. Weller’s group has suggested that 20 min of sun is mainly composed of collagen and elastic fibers [55] (Fig. 1). The
exposure can lower blood pressure for hours [50]. Solar radiation hypodermis was long considered to be part of the skin, though
activates the production of nitric oxide, a potent vasodilator, which it is currently viewed as part of a subcutaneous tissue with an
lowers blood pressure [51]. This may explain the higher incidence important influence on skin biology [59,60].
of cardiovascular disease at higher latitudes than in the tropics [52]. Several environmental factors pose a threat to skin physiology.
Exposure to visible light appears to stimulate an opsin that mod- In an important publication, Passeron’s group identified several
ulates the activity of subcutaneous white adipose tissue cells, the harmful factors for skin and coined the term “skin exposome”. The
main fat deposit in humans [53]. In addition, a study conducted by following factors are currently known to affect the biology of the
Lindqvist between 1990 and 2010 with 30,000 women indicated skin: 1) solar radiation (UV and infrared radiation, visible light),
that the average lifespan of women with regular sun exposure was 2) air pollution, 3) tobacco smoke, 4) nutrition, 5) miscellaneous
two years longer than those who did not receive much sunlight factors which have not been as thoroughly studied, such as stress,
[54]. Furthermore, sun-avoiding women had higher risks of heart sleep deprivation, temperature, and cosmetics [61].
problems, autoimmune diseases, and diabetes. Of these factors, this review will focus primarily on the effects
These results highlight the urgent need for a novel photoprotec- of UV radiation and visible light on the skin. UV radiation is
tion paradigm that considers the benefits of moderate sun exposure classified into UVC (100–280 nm), UVB (280–320 nm), and UVA
without sunscreen usage beyond the positive effects of vitamin (320–400 nm) (Fig. 2). These definitions were constructed at the
D. Moderate sunbathing with no sunscreen has many benefits turn of the 20th century though their classification has little to
for human welfare and health, and may even provide protection do with skin photobiology [58]. Since energy is inversely propor-
against skin cancer. tional to the wavelength, UVC has the highest energy, followed by
In order to understand the diverse and contradictory effects of UVB and UVA [57]. The skin penetration capacity of UV radiation is
skin exposure to sunlight, we need to start considering how light directly proportional to its wavelength: UVB reaches only the epi-
interacts with skin. In particular, we will pay special attention to the dermis and UVA reaches the dermis (Fig. 3, Table 1) [62–64]. The
reactions induced by light when absorbed by endogenous photo- UV radiation that reaches the Earth comprises only 2 % of the total
sensitizers and the consequences of the photosensitized oxidation solar radiation reaching the surface, of which UVA and UVB repre-
reactions for biomolecules and cells. We will also comprehensively sent approximately 90–95 % and 5–10 %, respectively, while UVC is

4
L.V.M. de Assis, P.N. Tonolli, M.N. Moraes et al. Journal of Photochemistry and Photobiology C: Photochemistry Reviews 47 (2021) 100403

Fig. 2. A. Spectrum of solar irradiance reaching the surface of the Earth: ultraviolet radiation UVB (200 – 320 nm) and UVA (320 – 400 nm), visible light (400 – 750 nm), and
infrared radiation (750 – 2500 nm). B. Absorption of mammalian opsins and main skin endogenous photosensitizers and biomolecules. Black line represents the range of
photon absorption through the electronic transition, which takes place when electrons are excited to a higher energy level, and red line by vibrational transition (starting at
750 nm). Figure created by the authors.

Fig. 3. Penetration of different fractions of sunlight in the human skin. Image taken from [104] under the terms of creative commons.

5
L.V.M. de Assis, P.N. Tonolli, M.N. Moraes et al. Journal of Photochemistry and Photobiology C: Photochemistry Reviews 47 (2021) 100403

completely blocked by the ozone layer [63,64]. In this review, we Table 2


Absorption and reflection at different wavelength ranges from UV to near infrared.
will use the abbreviation UVR when discussing studies that utilize
Photon absorption was determined by [105,106]. Values for reflection, scatter-
both UVA and UVB emission devices. ing, and thermal dissipation (represented in Fig. 1) were generated by subtracting
The link between chronic UVR exposure and skin cancer is well absorption values from 1.
documented in the literature [2]. The effects of UV radiation on
Wavelength (nm) Absorption Reflection
the skin include: I) sunburn, caused mainly by UVB [63,64]; II) /scattering/thermal
damage to nuclear and mitochondrial DNA resulting in the accu- energy dissipation
mulation of cyclobutane pyrimidine dimers and 6–4 photoproducts
UVB (280–320) 0.1 0.9
(UVB mainly), and of 8-oxoguanine (8-oxoG) due to the indirect UVA1 (340–400) 0.3 0.7
action of UVA [63,67,68]; III) DNA repair, which may trigger apo- UVA2 (320–340) 0.2 0.8
ptosis, resulting in skin shedding [64,69]; IV) generation of reactive Blue (450–480) 0.8 0.2
Green, Yellow, Orange (500–625) 0.7 0.3
species of oxygen and nitrogen (ROS and RNS), which damage lipids
Red (625–750) 0.6 0.4
and membranes [64,67,69]; V) activation of a complex neuroen- Near Infrared (750–1400) 0.5 0.5
docrine system in the skin with likely systemic effects [70,71];
VI) pre-vitamin D3 generation as a result of 7- dehydrocholes-
terol conversion by UVB [72]; VII) increased ␣-MSH production biological parameters with the external environment in a time-
that leads to immediate and delayed pigmentary responses [58]; coordinated fashion [96–98].
VIII) degradation of cellular matrix by metalloproteinases which is
associated with skin aging [62]; IX) immunosuppression associated 3. Upon absorption light photons generate excited states
with decreased immunosurveillance [58,63,64,67].
Visible light (400–750 nm) comprises approximately half of the The sun emits a broad spectrum of electromagnetic radiation
total solar irradiance reaching the Earth’s surface and is represented that includes gamma rays, ultraviolet radiation, and visible light
by several wavelengths, which penetrate the skin to different (the maximum intensity of solar radiation occurs at wavelengths of
extents, with longer wavelengths penetrating deeper. Visible light around 500 nm), as well as infrared, microwaves, and radio waves.
is comprised of the following shades, listed in decreasing order of However, a major part of this radiation does not reach the Earth’s
wavelength and skin penetration: red light (625–740 nm), orange surface. The main reason is that the Earth is protected by its mag-
(590–625 nm), yellow (565–590 nm), green (500–565 nm), blue netic field, the ozone layer, and by other atmospheric properties
(450–485 nm), and violet (400–450 nm) (Table 1, Fig. 3) [65,66]. that restrict the penetration of electromagnetic radiation [99]. The
Blue light is one of the most studied visible light wavelengths ozone layer absorbs radiation below 280 nm, meaning that only
and it has been implicated in the proliferation and differentia- UVA and UVB, visible, and infrared radiation (IRA, 760–1440 nm;
tion of human cells, as well as in the increased oxidative stress IRB, 1440–3000 nm, and IRC 3000 nm–10000 nm) will come into
of keratinocytes [73], systemic release of ␤-endorphin [71], and contact with human skin. As mentioned above, UV comprises only
hair follicle growth in ex vivo [74]. In murine models, blue light ∼2 % of the solar radiation that reaches the Earth’s surface (Fig. 2).
elicits an anti-inflammatory response [75], apoptosis of melanoma Most of the mutagenic activity of UV radiation is due to UVB pho-
cells [76], and vasorelaxation [77]. Violet light has been shown to tons being absorbed directly by DNA molecules. However, most of
downregulate markers of human keratinocyte differentiation [78]. the photons that reach the skin are in the visible (∼47 %) and IR
Other wavelengths, such as green light, display anti-inflammatory (∼51 %) ranges (Fig. 2).
effects on rats [79] and a stimulatory activity on angiogenesis and It is important to note that the effect of solar radiation on skin
myofibroblast differentiation in a rat model of burn injury [80]. Yel- depends on how photons interact with skin molecules. Especially
low light has been shown to inhibit melanogenesis and to induce relevant is the process called electronic excitation, which occurs
autophagy in human melanocytes [81]. Red light, the farthest- when a molecule absorbs a photon and is raised from a low-lying
reaching wavelength of the visible light spectrum, has been shown electronic state (called ground state) to another electronic state of
to enhance DNA repair activity [82] and proliferation [83,84]. A higher energy, and the specific characteristics (lifetime and reac-
growing body of literature has documented the role of visible light tivity) of the created excited state. Note that both UV radiation
in deleterious effects that may contribute and/or lead to skin can- and visible light induce electronic transitions (Fig. 2), though they
cer [7,8,62]. However, a link between visible light exposure and skin differ in terms of the molecules they interact with and the exact
cancer remains controversial. This debate is of major importance characteristics of the excited states they bring about. The impor-
to public health, since the majority of sunscreens do not protect tant question to answer is whether these transient excited states
against visible light. Meanwhile, an exciting and fast-growing field react during their short lifetime and with which molecule. One
named photobiomodulation, which uses low-intensity lasers, has should also consider that photons from the different regions of the
shown that light stimulation under controlled conditions improves sun radiation spectrum will have completely different penetration
several human disorders and diseases (reviewed in [85,86]). How- depth in the skin (Fig. 3).
ever, photobiomodulation applications in medical routines have A number of processes prevent photons from penetrating the
been delayed due to the lack of standard procedures and protocols skin. These include reflection, scattering, and absorption (Fig. 1,
in both basic and clinical studies [85,87]. Table 2) [100,101]. Note that UV and most IR radiation does
Upon stimulation by UV radiation and/or visible light, several not penetrate deep into the skin (Table 1) [102]. UVB radiation
complex biological mechanisms are activated in order to sustain barely reaches the basal layer and UVA radiation penetrates a few
skin homeostasis. It is beyond the scope of this review to explore micrometers into the dermis, while IRB and IRC only reach the
such mechanisms, though it bears mentioning that skin’s defensive first molecular layers of the epidermis (Table 1). Most IR is effi-
responses to solar radiation include the following mechanisms: I) a ciently absorbed by O H and C H bonds and quickly transformed
local neuroendocrine system with a systemic effect [56,88,89]; II) a into heat. Photons in the visible and IRA ranges penetrate most
melanin-producing system that protects cells against the effects of deeply into the skin because photons in this wavelength region
direct and indirect UV radiation [56,57]; III) apoptosis of severely are neither scattered nor absorbed effectively, spreading through-
damaged cells [90–92]; IV) DNA repair [93]; V) anti-oxidative stress out the dermis and even reaching the hypodermis (Tables 1 and 2,
system [92,94,95]; VI) the molecular clock, which acts as a tem- Fig. 3), resulting in a wide range of interactions with these tissues
poral controller of the above-mentioned systems, aligning several [103,104]. Thus, the penetration depth of photons is a determin-

6
L.V.M. de Assis, P.N. Tonolli, M.N. Moraes et al. Journal of Photochemistry and Photobiology C: Photochemistry Reviews 47 (2021) 100403

ing factor in the range of molecules and the consequent biological


effects on the skin.
The popularity of measures to avoid UV radiation via sunscreen
use is based on an understanding that UV photons are mutagenic
and carcinogenic. However, one should always remember that the
photons are not mutagenic by themselves, instead, photons par-
ticipate of photochemical reactions by raising molecules to their
excited states that chemically react with other molecules. We will
discuss the specific interactions between UV photons and skin in
greater detail below. Importantly, the exclusive focus on the role of
UV radiation has resulted in an inadvertent conception of photons
in the visible regions as being safe for the skin, which is not borne
out by scientific observations.
Many decades ago, Leff and Krinsky [107] demonstrated that
red light photons (∼610 nm) are mutagenic to microalgae (Euglena
gracilis). This is because the chlorophyll molecules in microalgae
absorb light at this wavelength. The same question posed to explain
the mutagenic effect of UV radiation can be repeated here: how Fig. 4. Formation of pyrimidine dimers (cyclobutane pyrimidine dimers and (6-4)
can visible light in the red spectral region be mutagenic? As men- photoproducts) in adjacent pyrimidines in DNA during exposure to UV, especially
UVB. Image obtained from [112] under the terms of creative commons.
tioned above, it all depends on the absorbing molecules and the
type of excited states that are formed, more than on the energy of
the photon itself. Chlorophyll molecules engage in photosensitized
oxidation reactions upon absorption of photons at certain wave- regions in the genome (hotspots), such as the CpG islands and
lengths. Humans do not have chlorophyll in the skin and indeed methylated CpG sequences (where 5-methylcytosine is deami-
red light seems to be safe for us [85,87], but the concept of red nated to thymine), where they cause transition mutations of C to
light being safe, just because it has less energy than UV, falls to the T, 5-methylcytosine to T, and CC to TT [108,109]. The CpG islands
ground. The simple and precise explanation is that red light is safe (sequences with 1 kb, on average, and rich in G + C) are usually
for us because we do not have chlorophyll in our skin. However, we found at the promoter region of some tumor suppressor genes
do have several other molecules that can engage in photosensitized such as TP53, which is why these mutations are commonly asso-
oxidation reactions (see further discussion below). ciated with skin cancer [110]. According to Pfeifer and colleagues,
Reactions of the excited states can be grouped into two main in human skin cancers, about 35 % of all mutations in the p53 gene
types. Direct events are purely photochemical, with molecules are transitions at 5"-TCG and 5"-CCG [108].
in excited states reacting among themselves or with neighbor- Note that all of this is not a consequence of the intrinsic and
ing molecules, while indirect photosensitization events involve isolated properties of UVB photons, but instead is a consequence of
the absorbing molecules (called photosensitizers), forming excited the absorption of UVB photons by nucleic acids. Although all nucle-
states that exchange energy or electrons with neighboring obases (adenine, cytosine, guanine, and uridine) can absorb UVB,
molecules and usually returning to the ground state. As mentioned only a few excitation events will lead to photoproducts [111,112].
above, sunlight photons of different wavelength regions will inter- AS mentioned above, an electronically excited pyrimidine adjacent
act with different molecules located in distinct subcellular niches to another pyrimidine will react to form either a CPDs or a 6-4 PPs
defining their biological effects (Fig. 2). Photons with lower wave- (Fig. 4) [111,112]. In other words, the photon needs to be absorbed
lengths (and higher energy) will interact with a larger number of by the pyrimidine base in order to induce the mutagenesis. The
molecules. For example, UVB photons interact with a myriad of mutagenic effect of visible light in microalgae occurs in an anal-
molecules (proteins, nucleic acids, lipids), including the genetic ogous fashion [107]. In this case, the absorbing molecules are not
material of living cells, located mainly in the basal layer and the DNA, but instead chlorophyll, a photosynthetic pigment present in
upper layers of the dermis. Upon absorbing a photon, the nitroge- the microalgae. The induced reaction is not a direct cycloaddition
nous bases of the DNA in these cells are brought into an excited between adjacent pyrimidine dimers, but instead, is a photosensi-
state. Most of these excited states quickly return the absorbed tized oxidation reaction, which occurs due to the formation of an
energy as heat, fully replenishing their respective ground state excited state after the absorption of photons (Fig. 5).
species. However, a small fraction of these events will favor the Photosensitizers (PS) depend on light absorption at the proper
formation of excited states that will engage in a direct photochemi- wavelength to allow the formation of excited states, which are
cal reaction called a cycloaddition reaction. Cycloaddition reactions intrinsically more reactive than ground states. During their tran-
occur when a photon excites a deoxythymidine that is in molecular sient lifetimes, the duration of which depends on the radiative
contact with another deoxythymidine, forming either cyclobutane and non-radiative deactivation routes, numerous reactions can
pyrimidine dimers (CPDs) or, to a lesser extent, pyrimidine (6-4) occur. Usually, the initial excited state formed after light absorp-
pyrimidone photoproducts (6-4PPs) (Fig. 4). These adducts distort tion is a singlet excited state (PS(S1)), with a lifetime measured
the DNA double helix, unwinding and kinking it, affecting the inter- in nanoseconds (Fig. 5). Consequently, PS(S1) do not generally
action with transcription factors, replication, transcription, and result in diffusion-dependent reactions unless these are generated
DNA repair. If the damage is not recognized and repaired by DNA- in physical contact with the reactant [113] or when reactants are
repairing enzymes, it can be maintained in the DNA, where it leads present at high concentrations. Triplet excited states (PS(T1)) usu-
to mutations. ally live much longer than PS(S1) and are therefore key elements
The acute effect of these DNA alterations is a strong inflamma- of photosensitized oxidation reactions. PS(T1) are typically formed
tory response also known as sunburn, in which the skin becomes by intersystem crossing (spin inversion in the excited state) from
reddish and painful to touch. UVB radiation is also known to cause the PS(S1), but can also be formed by the suppression of PS(S1)
long-term effects including a transition mutation in dipyrimidine excited states by molecular oxygen or by an energy transfer process
sequences containing cytosine, especially at 5"-TCG and 5"-CCG from other photosensitizers present in the molecular surroundings
[108]. These mutations occur more frequently in some specific (Fig. 5).

7
L.V.M. de Assis, P.N. Tonolli, M.N. Moraes et al. Journal of Photochemistry and Photobiology C: Photochemistry Reviews 47 (2021) 100403

Fig. 5. Mechanisms of photosensitized oxidation reactions involving the molecular oxygen (O2 ). The photosensitizer (PS) is a molecule capable to absorb light depending on
its specific absorption spectra. Once excited, the PS goes from its ground state PS(S0) to its singlet excited PS(S1), and through a process called intersystem crossing forms
triplet excited PS(T1) states. Other mechanisms can also form triplet species, like suppression of singlet excited state by ground state oxygen and energy transfer from other
photosensitizers. Both PS(S1) and PS(T1) can react directly with biomolecules, like carbohydrates, lipids, proteins or nucleic acids, via Type I contact-dependent reaction,
resulting in formation of radicals, like PS, capable to initiate other radical chain reactions. Photosensitizers can work as electron donors or acceptors, but in the scheme, we
only show the PS working as a photo-oxidant (electron acceptor). PS· (an anion radical derived from the PS) can react with oxygen forming anion radical super oxide (O2 .− ).
Generation of O2 .- can also occur by the direct electron donation from PS(S1) and PS(T1) to O2 (these pathways are not shown in the scheme). Since triplets live much longer
than singlets, triplets are the main agent to cause biological oxidation. Otherwise, PS (T1) can react with molecular oxygen 3 O2 , via the Type II reaction, generating singlet
• •
oxygen (1 O2 ). Both of these diffusive oxidant species (1 O2, O2 − ) will affect the redox homeostasis of cells and tissues but 1 O2, is much more reactive than O2 − [114]. Figure
taken from [114] with permission.

There are two main mechanisms of photosensitized oxida- Table 3


Relevant endogenous photosensitizers with their respective wavelength range of
tions, termed type I and type II, and PS(T1) are significant to both
light absorption (␭ range) and efficiency of singlet oxygen generation (S).
mechanisms. Type I reactions depend on the encounter of the
excited-state species with biological substrates. These reactions Molecule ␭ range (nm) S [reference]
usually start with an electron or hydrogen abstraction, leading to Pro-vitaminD2 270−280 0.8 [119]
radical chain reactions. Excited states are stronger oxidizing and Vitamin D3 270−280 0.2 [120]
reducing agents and both PS(S1) and PS(T1) can behave as elec- Vitamin B-12 320−380; 450−600 0.2 [121]
All-trans retinal 400−600 0.6 [122]
tron donors and acceptors. In the scheme of Fig. 5 we only mention
All-trans retinol 400−600 0.8 [123]
the direct photo-induced oxidations by both PS(S1) and PS(T1), Riboflavin 300−500 0.5 [124]
meaning that these species accept an electron from a biological FMN 300−500 0.6 [125]
substrate and form the one-electron reduced radical derived from MiniSOG (LOV) 300−500 0.03 [125]
SOPP (LOV) 300−500 0.19 [125]
the PS. We do not mention the possibility of photo-induced reduc-
Pterin 200−400 0.18 [126]
tions, in which both PS(S1) and PS(T1) donate an electron to oxygen Folic acid 200−400 < 0.02 [126]
to form the anion radical superoxide (O2 .-). Although there are Thymine 230−300 0.07 [127]
other species that could receive an electron from PS(S1) and PS(T1), Uracil 230−300 0.13 [127]
molecular oxygen is the most obvious one for thermodynamic and Adenine 230−280 0.03 [127]
Cytosine 230−300 0.02 [127]
kinetic reasons [114]. It is worth mentioning that these pathways
Phenylalanine 200−280 0.06 [128]
are usually not as prevalent as the type II reactions and O2 .- is not Tyrosine 200−300 0.17 [128]
as reactive as 1 O2 [114]. Tryptophan 200−320 0.06 [128]
Type II refers to the energy transfer reaction from the PS triplet BSA 200−320 0.04 [128]
Bacteriochlorophyll a 300−800 0.61 [129]
state to molecular oxygen (3 O2 ), generating singlet oxygen (1 O2 )
Bacteriochlorophyll b 300−800 0.68 [129]
(Fig. 5). This is the most prevalent oxidative pathway for molecules Bacteriopheophytin a 300−800 0.69 [129]
that form PS(T1), since PS(S1) is short-lived and the energy transfer Bacteriopheophytin b 300−800 0.61 [129]
reaction between PS(T1) and oxygen is usually diffusion-limited, Reaction Center 300−800 0.03 [130]
while the formation of PS(T1) by an electron transfer to oxygen is a R. sphaeroides
Lipofuscin 300−500 ∼0.1 [131]
minor product [114]. 1 O2 is a strong oxidant and can add to the dou-
Melanin 300−800 ∼0.02 [17]
ble bond of lipids, proteins, and nucleic acids, starting peroxidation Urocanic 200−320 ∼0 [116]
reactions [114]. As shown by Baptista’s group, irreversible biolog-
ical damage occurs with the abstraction of a hydrogen from the
double bonds of biomolecules [114,115]. In addition, they suggest which is much more difficult to be directly evaluated, especially in
that the fact that the irreversible damage occurs due to contact- biological systems. Since 1 O2 is formed when triplets are generated
dependent reactions indicates that the damage can be confined in the presence of oxygen the yield of singlet oxygen defines a min-
within a nanometric location from the PS [115]. imum limit for the yield of triplets and indicates that a molecule is
As described above, the formation of a triplet excited state is a PS.
key to triggering the photosensitized oxidation reactions. But, are Importantly, Table 3 demonstrates that 1 O2 generation is the
there relevant photosensitizers in the skin that can form triplets rule rather than the exception for PS molecules. Basically, it is very
upon light absorption? There is plenty [116,117]. Table 3 provides difficult to generate an excited state completely avoiding triplet
an overview of the range of wavelengths absorbed by important generation. Note that several amino acids and nitrogenous bases
biological molecules that are present in the skin and the yield of are efficient photosensitizers. Vitamins, in general, generate triplets
singlet oxygen generation. Besides being one of the oxidant species efficiently, meaning that these molecules are effective in caus-
that cause the photosensitized oxidative damage, 1 O2 generation ing photooxidative damage. Indeed, vitamins are the main species
can also be used to report the presence of triplet excited states, responsible for the photooxidative damage induced by UVA, caus-

8
L.V.M. de Assis, P.N. Tonolli, M.N. Moraes et al. Journal of Photochemistry and Photobiology C: Photochemistry Reviews 47 (2021) 100403

Fig. 6. Families of opsins and the corresponding coupled G-proteins in eukaryotes Figure taken from [134] with permission. In this review, we used the gene and protein
nomenclature for human and mice, which can be found at https://www.genenames.org/about/guidelines/ and http://www.informatics.jax.org/mgihome/nomen/gene.shtml,
respectively.

ing biological damage to others by generating 1 O2 and disrupting organisms to sense light of different wavelengths. Interestingly,
the general metabolic processes of skin cells due to their degrada- 11-cis retinal can be isomerized to all-trans retinal by thermo-
tion in over-exposed skin. Besides being photoactive, they are also activation, though this occurs only at a very low rate in the
redox active and can become oxidized relatively easily. In fact, pho- mammalian retina [133].
todegradation of vitamins in skin cells is likely to be responsible for In all opsins, photosensitivity results from the photoisomeriza-
their metabolic failure and a major component of photoaging [118]. tion of 11-cis retinal into all-trans retinal upon a photon capture.
Note that the absorbance of many of these molecules extends to the Molecular transformations are then initiated, forming a partially
visible region of the spectrum, meaning that visible light also causes stable Meta II opsin, whose conformation enables it to activate the
effects similar to those known to occur with UVA (UVB is different G protein. Vertebrate opsins are known to activate G␣i/o or G␣q,
because of the direct reaction in DNA, as described above). whereas the RRH and RGR do not interact with G proteins and can
Other pigments that accumulate in the skin, such as melanin bind to all-trans retinal [135,136].
and lipofuscin, are also known to generate 1 O2 . Baptista’s group For decades, opsins were associated with vision and irradiance
has shown that visible light generates similar mutagenic lesions to detection, while their discovery in non-ocular tissues was limited to
those caused by UVA radiation [8,18]. That is why the skin, even the non-mammalian pineal gland [137–139] and brain [140,141].
when protected by sunscreen, defends itself from the effects of In 1998, the scientific community was struck by the news that
visible light, promoting pigmentation and leading to a tanned skin. a novel opsin was found in the skin of the clawed frog Xenopus
laevis in melanin producing cells called melanophores, which was
why the new photopigment was given the name melanopsin [142].
4. How visible light and UV radiation interact with opsins Another surprise came from the realization that melanopsin was
more homologous to Octopus rhodopsin than to a typical verte-
4.1. Discovery of the opsins in the skin brate opsin, even sharing the same signal transduction cascade
as the fruit fly, Drosophila, rhodopsin. But a real breakthrough of
Opsins are 7-transmembrane G-coupled receptors made up the paradigm about mammalian photoreception was the discov-
of a protein moiety and the chromophore retinaldehyde, which ery by the same group [143] of melanopsin mRNA in the human
are present in organisms from archaea to metazoans [132]. It retinal ganglion cell layer. Axons of this subpopulation of ganglion
has been established that the presently known mammalian pho- cells project into the suprachiasmatic nuclei, the central biological
topigments arose from three family lines: 1) rhodopsins and clock, whose endogenous circadian oscillation is then adjusted by
cone opsins, pinopsin, VA-opsin, parapinopsin and encephalopsin the light/dark cycle information to exactly 24 h [144].
(ciliary opsins); 2) the RRH (retinal pigment epithelium-derived At the time, it was not known that opsins, including melanopsin,
rhodopsin homologue or peropsin) and RGR (retinal G-protein could be expressed in the mammalian skin. But soon after, in
coupled receptor) (photoisomerases); 3) the melanopsin line (rhab- 2001, Miyashita and colleagues [145] were the first to report the
domeric opsins) (Fig. 6). The protein residue provides the optimal expression of rhodopsin in the immortalized murine melanocytes
microenvironment for the absorption of light at a particular Melan-a, confirmed by Castrucci’s group [146], as well as in mouse
wavelength, and small differences near the chromophore allow skin, followed by reports of opsin expression and photoreception

9
L.V.M. de Assis, P.N. Tonolli, M.N. Moraes et al. Journal of Photochemistry and Photobiology C: Photochemistry Reviews 47 (2021) 100403

Table 4
Spectrum profile of mammalian opsins. Data was based on [15,150,172–175].

Range of detection (nm) Maximal absorption (nm) Reported to be sensitive to other wavelengths

OPN1SW 370 – 500 360 (mice) 420 (humans)


OPN1MW 400 – 650 510 (mice) 530 (humans)
OPN1LW 400 – 680 560 nm (humans)
OPN2 400 – 600 500 nm UVR (280–400 nm) and UVA radiation (365 nm)
OPN3 No evidence in mammals
OPN4 400 – 570 480 nm UVA radiation (365 nm)
OPN5 370 – 450 380 nm

by non-ocular tissues in non-mammalian vertebrates [147,148]. lent in-depth reviews [161,162]. As detailed below, the cis-retinal
It took almost another decade before opsins were discovered in undergoes substantial changes in its microenvironment during and
human skin [149]. Although their functions remained elusive, just after light absorption, as indicated by robust changes in its
immunolabeling of rhodopsin-OPN2 and of S and L/M cone opsins- absorption spectrum.
OPN1 was demonstrated in the human epidermis [149]. Wicks and The detailed steps of these transformations are probably the
colleagues [150] were able to confirm the expression of rhodopsin, best-studied examples of the conversion of light to mechanical
but none of the other opsins (encephalopsin-OPN3, melanopsin- energy. At every step, the process raises intriguing questions and
OPN4, neuropsin-OPN5) in human melanocytes, in contrast to the provides elegant solutions from nature. When the chromophore
findings in hairless mouse [151] and murine epidermal cells and absorbs a photon, it isomerizes from the 11-cis-retinal, which has
human skin [152], where the Opn5 gene and protein expression a bent shape, to the all-trans unbent form by the protonation
were detected. In a subsequent article, however, Oncea’s group [43], of a Schiff base, delocalizing ␲-electrons, and undergoing a red-
contradicting its previous report [150], demonstrated the presence shift absorption from 500 nm to 540 nm (bathorhodopsin form)
of OPN1SW, encephalopsin-OPN3 and neuropsin-OPN5 in human [158,163]. These conformational changes trigger a rearrangement
melanocytes and keratinocytes. Confirming the conspicuousness in the protein structure, causing allosteric interactions involved in
of rhodopsin transcripts in human skin, its expression was also the phototransduction responses.
demonstrated in human keratinocytes [78] and hair follicles [74]. Given the abundance and variability of the properties of opsins,
Castrucci’s group was the first to show the mRNA and pro- an abundance of questions surrounds the photophysics of opsins.
tein levels of melanopsin Opn4 in murine Melan-a and B16-F10 Why, for example, do most opsins function through the absorp-
cells [153], as well as confirmed the rhodopsin-Opn2, and in in tion of visible light, since retinal itself has an absorption maximum
silico analysis of human skin cone opsins OPN1SW, OPN1MW, in the UV region? Interestingly, the answer to this question not
and OPN1LW, rhodopsin-OPN2, encephalopsin-OPN3, melanopsin- only clarifies the red shift experienced by the chromophore itself,
OPN4, and neuropsin-OPN5 [15] in exposed and unexposed skin. but also explains why opsins are adapted to respond to such a
In addition, Toh and colleagues [154] found the peropsin gene and diverse range of wavelengths [158]. Inside rhodopsin, OPN2, the
protein in human skin and keratinocytes. In 2019, in human dermal apoprotein retinal binds to K296 in helix VII, forming a protonated
fibroblasts, mRNA and protein levels of OPN1, OPN2, OPN3, OPN4, Schiff base, increasing electron delocalization within the retinal
and OPN5 were detected [155]. Immediately after, OPN4 expression electronic configuration and, consequently, shifting its absorption
was also demonstrated in human keratinocytes and melanocytes spectrum towards visible light. A positive charge inside the protein
[156]. Importantly, RPE65, the isomerase essential for the regener- structure produces a large positive Gibbs energy potential, which is
ation of 11-cis retinal from all-trans retinal has also been found in reduced by the formation of an ion pair with a negatively charged
human keratinocytes [157]. amino acid residue. Fine tuning of the distances and strengths of
The functionality of these opsins in the skin as photosensors of these interactions allow shifts in the absorption spectra in the hun-
visible and UV radiation will be discussed in the following sections. dreds of nanometers, explaining why opsins work in such a wide
range of wavelengths [164]. As a consequence, the retinal itself has
4.2. Photophysical properties of opsins a maximal absorption in the UVR (380 nm), but this absorption is
shifted to other wavelengths, as shown in Table 4.
As already mentioned, in terms of their structure, opsins belong After light absorption, the major photochemical event is a pho-
to a family of transmembrane proteins, containing seven ␣-helix toisomerization reaction from the 11-cis retinal to the all-trans
with a prosthetic photoactive group, which is called the retinal (an form. Several intermediates have been identified during rhodopsin
aldehyde-form of vitamin A). Retinal, which exists in four different photocycle within timescales varying from picoseconds to mil-
forms, the more prevalent A1 (retinal) and A2 (3,4-dehydroretinal, liseconds [163–165]. The large quantum yield (0.65) [166] and
commonly found in vertebrates [158], and the less common A3 (3- the velocity of the photoisomerization (the primary ground-state
hydroxyretinal) and A4 (4-hydroxyretinal), is covalently bound to rhodopsin photoproduct occurs within 200 fs [167]), has been
a lysine residue at helix 7 and associated with G protein-coupled intriguing the scientific community for quite some time. The
receptors [158,159]. detailed mechanism for this one-way, extremely fast and efficient
Opsins enable the transformation of photons into potential photoisomerization reaction was initially proposed theoretically
energy that drives mechanical changes in the photoreceptor, which and recently proven experimentally to be the consequence of the
are ultimately transduced to an intracellular G protein. Bacteri- crossing between the potential energy surfaces of ground and
orhodopsin from Halobacterium halobium was the first isolated excited electronic states, in a so-called conical intersection [168].
and studied opsin. After light absorption (peak at 570 nm), bac- The regeneration of 11-cis retinal is processed with the help of the
teriorhodopsin is able to couple the cis-trans isomerization of the RPE 65, well known to catalyze this conversion in the retinal pig-
retinal group with conformational changes in the protein, transfer- ment epithelium in the mammalian eye [169]. Unlike the majority
ring protons from cytosol to the inter-membrane space and actively of the mammalian opsins, melanopsin OPN4 is a bistable photopig-
transforming light into a proton gradient, which can be used for ATP ment [170], meaning that it may exist as a mixture of two steady
synthesis [160]. It is beyond the scope of this review to explore the states: the 11-cis resting form and the all-trans excited form. This
role of opsins in microbial, and thus, the reader is referred to excel- photopigment may exhibit the ability to regenerate the 11-cis reti-

10
L.V.M. de Assis, P.N. Tonolli, M.N. Moraes et al. Journal of Photochemistry and Photobiology C: Photochemistry Reviews 47 (2021) 100403

nal without the need of an isomerase, similar to what happens in was dependent on the transient receptor potential ankyrin 1 chan-
invertebrates, where longer light wavelengths can turn all-trans nel (TRPA1), PLC, and calcium. Furthermore, UVR-induced early
back into 11-cis retinal [171]. melanin synthesis was also dependent on TRPA1 [175]. Interest-
Even though the thermal induced cis-trans isomerization has ingly, UVR was shown to transiently depolarize the membrane of
a very high energy barrier in the visual pigments, some opsins human melanocytes, which delayed TRPA1 inactivation, leading to
can also respond to temperature changes, with light-independent sustained calcium levels [184].
roles such as temperature discrimination [176]. This is intrigu- Concomitantly, UVA (10–100 kJ/m2 ) and violet light
ing, considering that the structure of classical opsins is designed (100–500 kJ/m2) exposure was shown to increase OPN2 tran-
to avoid electrical dark-noise signals [177]. However, there are scripts and downregulate differentiation markers in human
conditions that can lead to a decrease in the thermal activation keratinocytes. Over- or down-expressing OPN2 mRNA, resulted in,
barrier. For example, the deprotonation of the 11-cis Schiff base respectively, a decrease or increase in differentiation markers [89].
decreases the energy barrier of the cis-trans isomerization from ∼45 In 2014, Oancea’s group demonstrated that UVR activates a protein
to 27 kcal/mol. Therefore, if the pH of the opsin vicinity is slightly G␣q/11 dependent pathway that leads to PLC activation and
elevated, opsin deprotonation can occur, decreasing the energy bar- hydrolysis of phosphatidylinositol (4,5)-bisphosphate (PIP2 ) into
rier for thermal activation, whereupon the receptor may start to diacylglycerol (DAG) and inositol 1, 4, 5-trisphosphate (IP3 ). PIP2
respond to subtle temperature variations [178]. Recent evidence regulates TRPA1- dependent photocurrents (extracellular calcium
points to the opsin thermal activation being highly dependent on influx) while IP3 stimulates calcium release from intracellular
the entropy-driven breakup of the hydrogen bonding networks stores. The combination of TRPA1-mediated calcium entry and
(HBNs) that usually hinder the thermal isomerization reaction. calcium release from internal storage leads to sustained calcium
Indeed, rhodopsin mutants that have disrupted HBNs in the vicinity levels, which resulted in melanin synthesis [184].
of the 11-cis retinyl have significantly decreased thermal activa- Interestingly, mouse and human OPN5 have been demonstrated
tion energy [179]. This explains earlier observation that the thermal to be sensitive to UVR irradiation (380 nm [152]), and human
sensor in Drosophila larvae depends on the expression of different OPN5 expressed in HEK 293 cells responded to light (470−490 nm,
rhodopsins [180]. The thermosensing ability of opsins in mammals 2.5 × 10−8 kJ/m2 ) with transient calcium rise and to UVR radiation
has been explored in recent studies, though it remains elusive. (352–380 nm, 3.7 × 10-4 – 1.1 10-3 kJ/m2 ) with calcium rise, cAMP
The first report of mammalian opsins sensing thermal energy was production, and MAP kinase activation [185].
reported in pioneering work by Eisenbach’s group [181]. In these In 2017, Castrucci’s group explored the effect of UVA radiation
seminal reports, sperm cell thermotaxis was shown to be defective in association with heat shock on murine melanocytes. UVA radi-
in Opn2 or Opn4 knockout mice and dependent on phospholipase ation (4.4 kJ/m2 ) was shown to increase Opn4, Clock, and Bmal1
C (PLC) and cyclic nucleotides signaling pathways [181,182]. In transcripts in malignant melanocytes compared to unexposed cells.
skin cells, Castrucci’s group was also pioneer in demonstrating that Moreover, UVA radiation led to a transient increase in melanin lev-
OPN4 acts as a thermosensor in normal and malignant melanocytes, els, as a result of IPD processes. It should be emphasized that all
which feeds the local circadian clock with environmental temper- UVA-related data explained above was carried out at constant tem-
ature [183]. perature (37 ◦ C). Surprisingly, the authors found that when UVA
Collectively, as described above, there are solid photochemical- radiation was associated with temperature increase (up to 40 ◦ C) all
related mechanisms that provide the foundation for the thermo- UVA-induced responses were abolished, demonstrating that tem-
activation of opsins, which is followed by functionally driven perature could antagonize UVA-dependent effects [186]. This is
reports that have validated such concepts in in vitro and in vivo an important factor which has been neglected in most reports,
experiments. Therefore, opsins have been confirmed to sense heat since the majority of the literature ignores the temperature com-
as well as light. ponent in photobiology experimental designs. These data highlight
the importance of controlling temperature during UV/visible light
4.3. Advancement in the field: a historical view of opsin functions experiments since opsins can also act as a temperature sensor.
in the skin The process of melanogenesis within skin melanocytes and
the transfer of melanin granules to the adjacent keratinocytes are
4.3.1. UVR sensor important defensive mechanisms against UV radiation. Hu and col-
In 2010, Castrucci’s group reported the first mechanism of OPN2 leagues [187] reported that UVA (30 kJ/m2 ) and UVB (200 kJ/m2 )
signaling in a murine melanoma cell line, B16-F10. The authors elicited intracellular calcium rise in human melanocytes only in
demonstrated that Opn2 transcripts were increased by endothelin, the presence of retinal, which led to melanosome transfer in a
and that PLC, calcium, calcium/calmodulin kinase II, and protein co-culture of melanocytes and keratinocytes. This process was
kinase C were involved in the process [146]. enhanced when the cell culture was subject to simultaneous UVA
In 2011, Wicks and colleagues demonstrated that UVR (2 kJ/m2 , and UVB irradiation, and calcium influx was abolished when TRPM1
[150]) evoked a retinal-dependent calcium transient in human channel was silenced. Nevertheless, despite similar efficiency of
melanocytes. Calcium rise was shown to be mainly dependent on UVA and UVB in activating melanosome transfer, UVA is less effec-
UVA wavelength and intracellular calcium storages, G-protein sig- tive to stimulate melanin synthesis compared to UVB radiation.
naling and PLC. Only OPN2 transcript was detected in this study, Although UVB radiation leads to more significant DNA damage, it
and UVA-induced calcium increase was markedly reduced when it also activates protective mechanisms such as melanin production
was silenced. Moreover, a higher UVR dose (40 kJ/m2 ) led to a tem- [187].
poral increase of melanin levels, which was associated with UVA Castrucci’s group, using pharmacological and gene silencing
and retinal requirements. This was the first study to suggest that strategies, demonstrated that OPN4 is essential for UVA-induced
immediate pigment darkening (IPD) might be the result of de novo IPD in murine melanocytes. Moreover, UVA signaled through a
melanin synthesis [150], opposing a canonical view of IPD which pathway dependent on calcium, calcium/calmodulin kinase II,
had hitherto been associated with the oxidation of pre-existing nitric oxide (NOS), and cyclic guanosine monophosphate (cGMP).
melanin [58]. In a subsequent study, the same group [175] elegantly Interestingly, silencing Opn2 also abolished IPD, thus showing a
demonstrated that UVR elicits a retinal-dependent increase in ionic possible compensatory mechanism between OPN2 and OPN4 in
current in human melanocytes, which was mainly dependent on both normal and malignant melanocytes [15,153]. It should be
UVA but not UVB wavelength. The UVR-dependent photocurrent highlighted that in this experimental model, UVA induced a tran-

11
L.V.M. de Assis, P.N. Tonolli, M.N. Moraes et al. Journal of Photochemistry and Photobiology C: Photochemistry Reviews 47 (2021) 100403

sient rise in melanin levels, and not a sustained increase as observed 4.3.2. Visible light sensor
by Oncea’s group [150,174,175,184]. The reason for such difference Murine cell experiments by Castrucci’s group have also led to
may lie in different characteristics of the UV lamps, contami- advances in the knowledge of the physiological mechanisms reg-
nation with UVB, difference in irradiance, and cellular model as ulated by the skin photosensitive system. When these cells were
extensively discussed elsewhere [15,153,186]. A year later, blue kept in constant dark, twenty-four hours after visible light stimu-
light radiation (20 kJ/m2 ) was reported to reduce DNA synthesis lation (white light 0.85 kJ/m2 at constant temperature) a reduction
and stimulate differentiation in primary human epidermal ker- of Opn2 expression was seen in malignant melanocytes (B16-F10).
atinocytes. Upon OPN3 gene silencing, a slight decrease in the Meanwhile, 36 and 42 h after the same stimulus, an increase of
differentiation markers was found, which suggests that OPN3 may Opn4 mRNA was also observed in malignant cells, compared to
be required for barrier function in vivo. The role of OPN1SW was not controls kept in constant dark. No effects were detected in nor-
evaluated in this study, but its role as a putative blue light receptor mal melanocytes (Melan-a). OPN2 and OPN4 protein was detected
in this model was not discarded [188]. UVA radiation (100 kJ/m2 ) by immunocytochemistry and imaging flow cytometry in both cell
increased the protein levels of OPN1, OPN3, and OPN5 without lines, and upon light stimulation OPN4 migrated from the per-
leading to DNA damage in human fibroblasts. Upon knockdown of inuclear region to the membrane only in malignant melanocytes
OPN3, the UVA-induced increase of calcium level, the phosphory- [153]. Twenty-four hours after visible light stimulation, Per1, Per2,
lation of CREB and the activation of calmodulin-dependent protein and Clock expression was increased compared to unexposed malig-
kinase were abolished, along with the matrix metalloproteinases. nant melanocytes, while no effect was found on the clock genes
Thus, OPN3 was suggested to be the major player in UVA-elicited of normal melanocytes. In response to visible light, melanin lev-
photoaging [189] although no screening of the other opsins was els were unaffected in both cellular types [153]. Collectively, the
performed. Most studies in the literature show the involvement above-mentioned data in the murine model demonstrated that a
of OPN3 as a light/UVA sensor using gene silencing and pharma- functional opsin system was expressed in melanocytes, and under
cological strategies. However, a recent study by Oancea’s group these experimental conditions, seemed to be more sensitive in
has provided insights into a light-independent role of OPN3. In an malignant than in normal melanocytes [153].
elegant paper, OPN3 was ruled out as the melanocyte blue light In human keratinocytes, violet light (380 nm, 12 kJ/m2 ) induced
receptor using spectroscopy analysis, i.e., OPN3 did not absorb in the highest level of intracellular calcium, which was inhibited when
any UV/visible wavelength, despite its ability to bind to retinal [44]. retinal was withdrawn from the medium or a broad protein G
It should be noted that the first attempt to characterize mouse and inhibitor (suramin) was added. In addition, upon peropsin gene
human OPN3 was performed in 2016, but it was not successful silencing, the violet light-induced calcium flux was lost [154].
[172]. These findings demonstrated the inability of OPN3 to absorb In an interesting study using human hair follicles, blue light
photons, thus ruling it out as a light sensor, despite its ability to bind (453 nm, 32 kJ/cm2 ) was shown to prolong the anagen phase in
to retinal. Along the same lines, OPN3 was shown to be a negative an ex vivo model, which was associated with sustained prolif-
regulator of melanogenesis in human melanocytes, independently eration, while red light, at the same dose, did not affect hair
of UV and visible light, by inhibiting a MC1R (melanocortin type growth. OPN3 gene silencing in the hair follicles decreased the
1 receptor)-dependent cAMP pathway [44]. A more recent report majority of proliferation- and apoptosis-related genes as well as
has also brought more evidence of a light and thermo-independent abolishing the blue light-induced effects [74]. In melanocytes from
role of OPN3. In human melanocytes, OPN3 gene knockdown led phototype IV human individuals, blue light (415 nm, 500 kJ/m2 )
to apoptosis in a calcium-dependent manner, with increased mito- increased OPN3- dependent pigmentation [14]. Unlike a previous
chondria permeability and caspase activation [190]. report [43], Passeron’s group found no opsins but OPN3 in human
In 2020, Castrucci’s group demonstrated a novel and interest- melanocytes and that blue light stimulation did not affect its tran-
ing role of OPN4. With three daily low doses of UVA radiation script levels. However, blue light triggered a signaling pathway
(4.4 kJ/m2 ), a reduction on cellular proliferation was found in which involved increased calcium levels, activation of calmodulin-
normal and malignant melanocytes; however, cell death, in an dependent protein kinase II, phosphorylation of CREB, extracellular
apoptosis-dependent manner, was only found in the latter group. signal-regulated kinase, p38, upregulation of microphthalmia-
Daily doses of UVA also led to persistent pigment darkening (PPD) associated transcription factor (MITF), and increased expression
in both cell types. Interestingly, upon Opn4 gene knockout by Clus- of the melanogenic enzymes, tyrosinase, and dopachrome tau-
tered Regularly Interspaced Short Palindromic Repeats (CRISPR) tomerase. Upon OPN3 silencing, blue light-induced signaling
technique, all UVA-induced effects were lost. In this study, an cascade, described above, was suppressed, and consequently no
increased molecular clock activation was found in Opn4 KO cells melanin increase was seen. Interestingly, this study also ruled out
compared to wild type counterparts, which suggests a putative the oxidative stress as a participant in this process, thus strength-
compensatory mechanism by other opsins, such as OPN2, which ening the evidence of an opsin-based detection system [14].
could lead to increased responsiveness. Moreover, bioinformat- Also in 2019, Buhr and colleagues demonstrated OPN5 expres-
ics analysis revealed that OPN4 expression gradually decreases sion in mouse vibrissal pad and ear pinna skin. Under light/dark
with human melanoma progression [191]. Therefore, this study cycle, the skin failed to be synchronized to the light-dark cycle
provided initial evidence of OPN4 functions in PPD and in UVA- in the absence of retinal as well as in the Opn5−/− mice. Another
induced apoptosis, as well as a putative role in the carcinogenic evidence of OPN5-neuropsin function arose from ex vivo skin exper-
process of cutaneous melanoma [191]. More recently, Oancea’s iments kept in complete darkness and exposed to 27 kJ/m2 of violet
group demonstrated that UVA-induced ROS generation leads to light (at 36 ◦ C). Such light stimulus entrained the molecular clock
melanin synthesis through two different temporally distinct mech- entrainment, which was abolished in the skin from Opn5−/− mice.
anisms. The early process is upstream the UVA-induced signaling Interestingly, the skin molecular clock of blind mice, with rod and
cascade activation. The second one requires retinal and G␣q/11- cone degeneration associated with OPN4-melanopsin deletion, was
dependent calcium mobilization into the mitochondria. Therefore, synchronized to the light/ dark cycle which together with the pre-
this study demonstrated a novel evidence that mitochondria of vious data demonstrate that the skin can directly sense light, and
melanocytes are able to respond to UVA radiation effects, contribut- this is dependent on OPN5, and not OPN4 [193].
ing to melanin synthesis; however, the identity of the opsin that Fan and colleagues elegantly showed that daily blue light
participates in UVA phototransduction cascade, in this particular (463 ± 50 nm, 20 kJ/m2 ) for 10 days stimulated hair follicle anagen,
model, remains elusive [192]. leading to a significant hair growth compared to unexposed ani-

12
L.V.M. de Assis, P.N. Tonolli, M.N. Moraes et al. Journal of Photochemistry and Photobiology C: Photochemistry Reviews 47 (2021) 100403

mals. Green light stimulation (522 ± 50 nm), at the same dose, also with its different cell types, the complex communication between
stimulated hair growth but less effectively. Blue light-induced hair its cells, the signaling pathways and biological processes that take
growth was lost when the optic nerve was crushed and reduced in place in this organ. A significant advancement in the photobiol-
animals with rod and cone degeneration. Interestingly, in Opn4−/− ogy of the skin has been achieved by the hard work of several
mice, blue light stimulation did not affect hair growth compared groups worldwide. To further unravel more layers of knowledge
to wild type animals. Mechanistically, blue light stimulation to the in this system, we understand that a more integrative approach is
eyes increased systemic sympathetic activity and norepinephrine needed. In other words, the mechanism of photons’ interaction with
levels in the skin, and activated hedgehog pathways in wild type chromophores and opsins must be addressed in future research
animals. All these effects were lost in Opn4−/− animals [194]. projects. Nonetheless, the contribution of previous studies focus-
In 2020, Kusumoto and colleagues reported OPN4 expression ing the theme cannot be disregarded. On the contrary, it must be
in human skin tissue and in primary melanocytes, keratinocytes, acknowledged!
and fibroblasts. A new variant of Opn4 was detected, but it was Due to space limitation, we could not properly give credit to
considered non-functional due to the lack of retinal binding site. In several important studies that laid the foundation of our current
human fibroblasts kept in dark, blue light stimulation (450–488 nm, knowledge. The understanding of how light is detected by skin
10–30 ␮mol m−2 s−1 during 0–20 min), applied in cells kept at cells might be dependent on mutual interaction between opsins
37 ◦ C, led to calcium influx and ERK1/2 phosphorylation, which and chromophores, which itself is a challenge to be experimen-
were reduced when OPN4 was antagonized with opsinamide [156]. tally addressed. Within this line, our goal was to provide a critical
The inability of OPN3 to absorb UV/visible light photons thinking to the reader that UV and light photons can interact simul-
reported by Ozdeslik and colleagues [44] calls for a deeper investi- taneously with both skin opsins and chromophores, as well as the
gation into the literature as several studies, described above, have benefits and the harm resulting from the exposure. We hope the
demonstrated that the light-induced effects were lost when the reader may see the skin as a complex light-detecting organ with
opsin expression was reduced. However, the loss or reduction of several phenomena taking place driven by photon stimulation. A
biological effects upon reducing OPN3 levels does not guarantee its better comprehension of such processes will increase our knowl-
function as a light sensor. One may suggest that OPN3 may partic- edge of skin biology as well as improve the therapy of skin-related
ipate as a player in the signaling cascade but not as a sensor. Or diseases. The future is promising.
considering that over the past decades, opsins have been shown to
be intriguing molecules: they were historically seen as light sensors Declaration of Competing Interest
but were shown to act as thermosensor, and, more recently, opsins
were reported to be involved in taste discrimination in Drosophila All authors state no conflict of interest that could have impacted
[195]. Therefore, it would not be surprising for opsins to also per- the development of this study.
form unexpected roles as GPCRs. Another important feature of
OPN3 is its wide distribution in several “blind” tissues, which do not
receive light, thus arguing for light-independent effects, despite its Acknowledgements
canonical ability to bind to retinal.
The caveats of pharmacological and gene silencing techniques This work was supported by the Sao Paulo Research Founda-
must be considered. Caution must therefore be used when inter- tion (FAPESP, grants 2017/24615-5 and 2018/14728-0 to Castrucci
preting experiments and calling an opsin a light sensor. Such AML; CEPID REDOXOMA 2013/07937-8 to Baptista MS) and by
confusion has been seen in the rapid effects of aldosterone. In the National Council of Technological and Scientific Development
recent years, several experiments showed that G-protein coupled (CNPq grants 303078/2019-7 to Castrucci AML; 303831/2019-7
estrogen receptor 1 (GPER1) was necessary for inducing the rapid to Baptista MS). Moraes MN is a Young Investigator of FAPESP
effects of aldosterone. Those experiments were mainly based on (2017/26651-9). de Assis LVM was a fellow of FAPESP (2018/16511-
gene silencing and pharmacological strategy, similarly to OPN3- 8). Funding sources had no involvement in the study design, in the
related experiments. In fact, the absence of GPER1 led to abolish- collection, analysis and interpretation of data, in the writing of the
ment of many aldosterone-induced effects, and as consequence, report, and in the decision to submit the article for publication.
GPER1 received attention as a new receptor for aldosterone. How-
ever, further investigation has demonstrated that aldosterone does References
not bind GPER1 in the membrane (review in [196]). As is the case
of aldosterone and its inability to bind to GPER1, may be analo- [1] U. Leiter, T. Eigentler, C. Garbe, Epidemiology of skin cancer, Adv. Exp. Med.
Biol. 810 (2014) 120–140, http://dx.doi.org/10.1007/978-1-4939-0437-2 7.
gous to the inability of OPN3 to be activated by light according [2] L. Cohen, M.A. Brodsky, R. Zubair, I. Kohli, I.H. Hamzavi, M. Sadeghpour,
to spectroscopy data [44]. Indeed, OPN3 may participate in the Cutaneous interaction with visible light: what do we know, J. Am. Acad.
blue light-induced pathway, as it has been clearly shown in several Dermatol. (2020), http://dx.doi.org/10.1016/j.jaad.2020.03.115.
[3] AAD (American Academy of Dermatology), Vitamin D and UV exposure,
studies; however, its role as the effective sensor remains unclear. (n.d.). https://www.aad.org/media/stats/prevention-and-care/vitamin-d-
Taken together, since the first report in 2001, and especially and-uv-exposure.
after 2011, the knowledge of the photosensitive system of the [4] Z. Apalla, A. Lallas, E. Sotiriou, E. Lazaridou, D. Ioannides, Epidemiological
trends in skin cancer, Dermatol. Pract. Concept. 7 (2017) 1–6, http://dx.doi.
skin has seen a significant advancement as many opsin-dependent org/10.5826/dpc.0702a01.
skin physiological processes and putative targets were discovered [5] TMR (Transparency Market Research), Sun Care Market (Type - SPF 6-14,
(Fig. 1), (Graphical Abstract). Still, there are many unknowns and SPF 15-30, SPF 30-50, SPF 50+; Form - Creams, Gel, Lotion, Powder, Liquid,
Wipes, Spray, Colored) - Global Industry Analysis, Size, Share, Growth,
open questions. With gene editing and omics approaches, new dis-
Trends and Forecast 2016-2024, 2015.
coveries will be made and putative drug targets for skin-related dis- [6] L. Kolbe, How much sun protection is needed?: Are we on the way to
eases will be revealed and translated into clinical care treatments. full-spectrum protection? J. Invest. Dermatol. 132 (7) (2012) 1756–1757,
http://dx.doi.org/10.1038/jid.2012.148.
[7] F. Liebel, S. Kaur, E. Ruvolo, N. Kollias, M.D. Southall, Irradiation of skin with
5. Concluding remarks visible light induces reactive oxygen species and matrix-degrading
enzymes, J. Invest. Dermatol. 132 (7) (2012) 1901–1907, http://dx.doi.org/
Understanding how UV radiation and light photons interact 10.1038/jid.2011.476.
[8] P.N. Tonolli, O. Chiarelli-Neto, C. Santacruz-Perez, H.C. Junqueira, I.S.
with the skin has been an ongoing challenge but a rewarding adven- Watanabe, F.G. Ravagnani, W.K. Martins, M.S. Baptista, Lipofuscin generated
ture. The beauty that lies within the skin tissue must be appreciated, by UVA turns keratinocytes photosensitive to visible light, J. Invest.

13
L.V.M. de Assis, P.N. Tonolli, M.N. Moraes et al. Journal of Photochemistry and Photobiology C: Photochemistry Reviews 47 (2021) 100403

Dermatol. 137 (11) (2017) 2447–2450, http://dx.doi.org/10.1016/j.jid.2017. [34] Z. Naeem, Vitamin d deficiency- an ignored epidemic, Int. J. Health Sci.
06.018. (Qassim) 4 (1) (2010) V–VI.
[9] E. Markiewicz, O.C. Idowu, Melanogenic difference consideration in ethic [35] S. Pilz, N. Verheyen, M.R. Grübler, A. Tomaschitz, W. März, Vitamin D and
skin type: a balance approach between skin brightening applications and cardiovascular disease prevention, Nat. Rev. Cardiol. 13 (2016) 404–417,
beneficial sun exposure, Clin. Cosm. Investig. Dermatol. 13 (2020) 215–232, http://dx.doi.org/10.1038/nrcardio.2016.73.
http://dx.doi.org/10.2147/CCID.S245043. [36] R. Civitelli, K. Ziambaras, Calcium and phosphate homeostasis: concerted
[10] M. Brenner, V.J. Hearing, The protective role of melanin against UV damage interplay of new regulators, J. Endocrinol. Invest. 34 (7) (2011) 3–7.
in human skin, Photochem. Photobiol. 84 (3) (2008) 539–549, http://dx.doi. [37] C. Aranow, Vitamin D and the immune system, J. Investig. Med. 59 (2012)
org/10.1111/j.1751- 1097.2007.00226.x. 881–886, doi:10.231/JIM.0b013e31821b8755.
[11] L. Zastrow, N. Groth, F. Klein, D. Kockott, J. Lademann, R. Renneberg, L. [38] S. Penckofer, J. Kouba, M. Byrn, C.E. Ferrans, Vitamin D and depression:
Ferrero, The missing link - Light-induced (280-1,600 nm) free radical where is all the sunshine, Issues Ment. Health Nurs. 31 (2010) 385–393,
formation in human skin, Skin Pharmacol. Physiol. 22 (1) (2009) 31–44, http://dx.doi.org/10.3109/01612840903437657.
http://dx.doi.org/10.1159/000188083, 2009. [39] A.J. Samanek, E.J. Croager, P. Gies, E. Milne, R. Prince, A.J. McMichael, R.M.
[12] S. Cho, M.H. Shin, Y.K. Kim, J.E. Seo, Y.M. Lee, C.H. Park, J.H. Chung, Effects of Lucas, T. Slevin, Estimates of beneficial and harmful sun exposure times
infrared radiation and heat on human skin aging in vivo, J. Investig. during the year for major Australian population centres, Med. J. Aust. 184
Dermatol. Symp. Proc. 14 (1) (2009) 15–19, http://dx.doi.org/10.1038/ (7) (2006) 338–341.
jidsymp.2009.7. [40] M.F. Holick, T.C. Chen, Vitamin D deficiency: a worldwide problem with
[13] S. George, M.R. Hamblin, H. Abrahamse, Effect of red light and near infrared health consequences, Am. J. Clin. Nutr. 87 (4) (2008) 1080S–1086S, http://
laser on the generation of reactive oxygen species in primary dermal dx.doi.org/10.1093/ajcn/87.4.1080S.
fibroblasts, J. Photochem. Photobiol. B, Biol. 188 (2018) 60–68, http://dx.doi. [41] W.B. Grant, C.F. Garland, E.D. Gorham, An estimate of cancer mortality rate
org/10.1016/j.jphotobiol.2018.09.004. reductions in Europe and the US with 1,000 IU of oral vitamin D per day,
[14] C. Regazzetti, L. Sormani, D. Debayle, F. Bernerd, M.K. Tulic, G.M. De Donatis, Recent Results Cancer Res. 174 (2007) 225–234, http://dx.doi.org/10.1007/
B. Chignon-Sicard, S. Rocchi, T. Passeron, Melanocytes sense blue light and 978-3-540-37696-5 20.
regulate pigmentation through opsin-3, J. Invest. Dermatol. 138 (1) (2017) [42] CDC, Skin Cancer Cases and Costs on the Rise from 2002-2011, US
171–178, http://dx.doi.org/10.1016/j.jid.2017.07.833. Department Health and Human Diseases, 2014 https://www.cdc.gov/media/
[15] L.V.M. de Assis, M.N. Moraes, K.K. Magalhaes-Marques, A.M.L. Castrucci, releases/2014/p1110-skin-cancer.html.
Melanopsin and rhodopsin mediate UVA-induced immediate pigment [43] K. Haltaufderhyde, R.N. Ozdeslik, N.L. Wicks, J.A. Najera, E. Oancea, Opsin
darkening:unravelling the photosensitive system of the skin, Eur. J. Cell Biol. expression in human epidermal skin, Photochem. Photobiol. 91 (1) (2015)
97 (3) (2018) 150–162, http://dx.doi.org/10.1016/j.ejcb.2018.01.004. 117–123, http://dx.doi.org/10.1111/php.12354.
[16] K.H. Kaidbey, P.P. Agin, R.M. Sayre, A.M. Kligman, Photoprotection by [44] R.N. Ozdeslik, L.E. Olinski, M.M. Trieu, D.D. Oprian, E. Oancea, Human
melanin—a comparison of black and Caucasian skin, J. Am. Acad. Dermatol. 1 nonvisual opsin 3 regulates pigmentation of epidermal melanocytes
(3) (1979) 249–260, http://dx.doi.org/10.1016/S0190-9622(79)70018-1. through functional interaction with melanocortin 1 receptor, Proc. Natl.
[17] O. Chiarelli-Neto, C. Pavani, A.S. Ferreira, A.F. Uchoa, D. Severino, Generation Acad. Sci. U. S. A. 116 (23) (2019) 11508–11517, http://dx.doi.org/10.1073/
and suppression of singlet oxygen in hair by photosensitization of melanin, pnas.1902825116.
Free Rad. Biol. Med. 51 (6) (2011) 1195–1202, http://dx.doi.org/10.1016/j. [45] N.Y. Leung, C. Montell, Unconventional roles of opsins, Annu. Rev. Cell Dev.
freeradbiomed.2011.06.013. Biol. 33 (2017) 241–264, http://dx.doi.org/10.1146/annurev-cellbio-
[18] O. Chiarelli-Neto, A.S. Ferreira, W.K. Martins, C. Pavani, D. Severino, F. Faião- 100616-060432.
Flores, S.S. Maria-Engler, E. Aliprandini, G.R. Martinez, P. Di Mascio, M.G. [46] J.F. Duffy, C.A. Czeisler, Effect of light on human circadian physiology, Sleep
Medeiros, M.S. Baptista, Photosensitization of melanin and the effect of Med. Clin. 4 (2) (2009) 165–177, http://dx.doi.org/10.1016/j.jsmc.2009.01.
visible light on skin and hair, PLoS One 9 (11) (2014) e113266, http://dx.doi. 004.
org/10.1371/journal.pone.0113266. [47] T. Roenneberg, T. Kantermann, M. Juda, C. Vetter, K.V. Allebrandt, Light and
[19] O. Chiarelli-Neto, M.S. Baptista, Photosensitizing properties of melanin upon the human circadian clock, Handb. Exp. Pharmacol. 217 (2013) 311–331,
excitation with visible light, Trends Photochem. Photobiol. 17 (2016) 57–68. http://dx.doi.org/10.1007/978-3-642-25950-0-13.
[20] N.A. Vashi, M.B.D.C. Maymone, R.V. Kundu, Aging differences in ethnic skin, [48] T. Roenneberg, C.J. Kumar, M. Merrow, The circadian clock and human
J. Clin. Aesthet. Dermatol. 9 (1) (2016) 31–38. health, Curr. Biol. 26 (10) (2016) R432–443, http://dx.doi.org/10.1016/j.cub.
[21] S.V. Davis, Vitamin D deficiency and type 2 diabetes in African Americans: 2016.04.011.
the common denominators, Diabetes Spectr. 24 (3) (2011) 148–153, http:// [49] N. Fleury, S. Geldenhuys, S. Gorman, Sun exposure and its effects on human
dx.doi.org/10.2337/diaspect.24.3.148. health: mechanisms through which sun exposure could reduce the risk of
[22] IARC 1992 IARC Monograph on the evaluation of carcinogenic risks to developing obesity and cardiometabolic dysfunction, Int. J. Environ. Res.
human. Solar and ultraviolet radiation, IARC Monogr. Eval. Carcinog. Risks Public Health 13 (10) (2016) 999, http://dx.doi.org/10.3390/ijerph13100999.
Hum. 55 (1992) 1–316, PMID 1345607. [50] D. Liu, B.O. Fernandez, A. Hamilton, N.N. Lang, J.M.C. Gallagher, D.E. Newby,
[23] M.S. Matsui, A. Hsia, J.D. Miller, K. Hanneman, H. Scull, K.D. Cooper, E. Baron, M. Feelisch, R.B. Weller, UVA irradiation of human skin vasodilates arterial
Non- sunscreen photoprotection: antioxidants add value to a sunscreen, JID vasculature and lowers blood pressure independently of nitric oxide
Symp. Proc. 14 (1) (2009) 56–59, http://dx.doi.org/10.1038/jidsymp.2009.14. synthase, J. Invest. Dermatol. 134 (7) (2014) 1839–1846, http://dx.doi.org/
[24] B. Eberlein-König, J. Ring, Relevance of vitamins C and E in cutaneous 10.1038/jid.2014.27.
photoprotection, J. Cosmet. Dermatol. 4 (1) (2005) 4–9, http://dx.doi.org/10. [51] G.M. Halliday, S.N. Byrne, An unexpected role: UVA-induced release of nitric
1111/j.1473-2165.2005.00151.x. oxide from skin may have unexpected health benefits, J. Invest. Dermatol.
[25] J.V. Freitas, H.C. Junqueira, W.K. Martins, M.S. Baptista, L.R. Gaspar, 134 (7) (2014) 1791–1794, http://dx.doi.org/10.1038/jid.2014.33.
Antioxidant role on the protection of melanocytes against visible [52] R.B. Weller, Sunlight has cardiovascular benefits independently of vitamin D,
light-induced photodamage, Free Rad. Biol. Med. 131 (2019) 399–407, Blood Purif. 41 (1-3) (2016) 130–134, http://dx.doi.org/10.1159/000441266.
http://dx.doi.org/10.1016/j.freeradbiomed.2018.12.028. [53] K. Ondrusova, M. Fatehi, A. Barr, Z. Czarnecka, W. Long, K. Suzuki, S.
[26] IARC, IARC handbooks of cancer prevention Sunscreens, Volume 5, 2001. Campbell, K. Philippaert, M. Hubert, E. Tredget, P. Kwan, N. Touret, M.
[27] L.K. Dennis, L.E.B. Freeman, M.J. VanBeek, Sunscreen use and the risk for Wabitsch, K.Y. Lee, P.E. Light, Subcutaneous white adipocytes express a light
melanoma: a quantitative review, Ann. Intern. Med. 139 (12) (2003) sensitive signaling pathway mediated via a melanopsin/TRPC channel axis,
966–978, http://dx.doi.org/10.7326/0003-4819-139-12-200312160-00006. Sci. Rep. 7 (1) (2017) 16332, http://dx.doi.org/10.1038/s41598-017-16689-4.
[28] R.P. Gallagher, Sunscreens in melanoma and skin cancer prevention, CMAJ [54] P.G. Lindqvist, E. Epstein, K. Nielsen, M. Landin-Olsson, C. Ingvar, H. Olsson,
173 (3) (2005) 244–245, http://dx.doi.org/10.1503/cmaj.050762. Avoidance of sun exposure as a risk factor for major causes of death: a
[29] J.C. DiNardo, C.A. Downs, Dermatological and environmental toxicological competing risk analysis of the melanoma in Southern Sweden cohort, J.
impact of the sunscreen ingredient oxybenzone/benzophenone-3, J. Cosmet. Intern. Med. 280 (4) (2016) 375–387, http://dx.doi.org/10.1111/joim.12496,
Dermatol. 17 (1) (2018) 15–19, http://dx.doi.org/10.1111/jocd.12449. 2016.
[30] FDA proposes changes to US sunscreen rules, in: C&EN Glob. Enterp., 2019, [55] E. McLafferty, C. Hendry, F. Alistair, The integumentary system: anatomy,
http://dx.doi.org/10.1021/cen-09709-polcon1. physiology and function of skin, Nurs. Stand. 27 (3) (2012) 35–42, http://dx.
[31] C.A. Downs, E. Kramarsky-Winter, R. Segal, J. Fauth, S. Knutson, O. Bronstein, doi.org/10.7748/ns2012.10.27.7.35.c9358.
F.R. Ciner, R. Jeger, Y. Lichtenfeld, C.M. Woodley, P. Pennington, K. Cadenas, [56] A. Slominski, D.J. Tobin, S. Shibahara, J. Wortsman, Melanin pigmentation in
A. Kushmaro, Y. Loya, Toxicopathological effects of the sunscreen UV filter, mammalian skin and its hormonal regulation, Physiol. Rev. 84 (4) (2004)
oxybenzone (benzophenone-3), on coral planulae and cultured primary 1155–1228, http://dx.doi.org/10.1152/physrev.00044.2003.
cells and its environmental contamination in Hawaii and the U.S. Virgin [57] J.Y. Lin, D.E. Fisher, Melanocyte biology and skin pigmentation, Nature 445
Islands, Arch. Environ. Contam. Toxicol. 70 (2016) 265–288, http://dx.doi. (7130) (2007) 843–850, http://dx.doi.org/10.1038/nature05660.
org/10.1007/s00244-015-0227-7. [58] L.R. Sklar, F. Almutawa, H.W. Lim, I. Hamzavi, Effects of ultraviolet radiation,
[32] R. Danovaro, L. Bongiorni, C. Corinaldesi, D. Giovannelli, E. Damiani, P. visible light, and infrared radiation on erythema and pigmentation: a
Astolfi, L. Greci, A. Pusceddu, Sunscreens cause coral bleaching by promoting review, Photochem. Photobiol. Sci. 12 (1) (2013) 54–64, http://dx.doi.org/10.
viral infections, Environ. Health Perspect. 116 (4) (2008) 421–572, http://dx. 1039/c2pp25152c.
doi.org/10.1289/ehp.10966. [59] G. Rivera-Gonzalez, B. Shook, V. Horsley, Adipocytes in skin health and
[33] State of Hawaii, 2018, S.B. 2571. Available on: https://www.capitol.hawaii. disease, Cold Spring Harb. Perspect. Med. 4 (3) (2014), http://dx.doi.org/10.
gov/session2018/bills/SB2571 CD1 .pdf. 1101/cshperspect.a015271.

14
L.V.M. de Assis, P.N. Tonolli, M.N. Moraes et al. Journal of Photochemistry and Photobiology C: Photochemistry Reviews 47 (2021) 100403

[60] I.L. Kruglikov, P.E. Scherer, Dermal adipocytes: from irrelevance to [83] S. Rohringer, W. Holnthoner, S. Chaudary, P. Slezak, E. Priglinger, M. Strassl,
metabolic targets? Trends Endocrinol. Metab. 27 (1) (2016) 1–10, http://dx. K. Pill, S. Muhleder, H. Redl, P. Dungel, The impact of wavelengths of LED
doi.org/10.1016/j.tem.2015.11.002. light-therapy on endothelial cells, Sci. Rep. 7 (1) (2017) 10700, http://dx.doi.
[61] T. Passeron, J. Krutmann, M.L. Andersen, R. Katta, C.C. Zouboulis, Clinical and org/10.1038/s41598-017-11061-y.
biological impact of the exposome on the skin, J. Eur. Acad. Dermatol. [84] H. Ma, J.-P. Yang, R.K. Tan, H.-W. Lee, S.-K. Han, Effect of low-level laser
Venereol. 34 (2020) 4–25, http://dx.doi.org/10.1111/jdv.16614. therapy on proliferation and collagen synthesis of human fibroblasts in
[62] E. Dupont, J. Gomez, D. Bilodeau, Beyond UV radiation: a skin under vitro, J. Wound Manag. Res. 14 (1) (2018) 1–6, http://dx.doi.org/10.22467/
challenge, Int. J. Cosmet. Sci. 35 (3) (2013) 224–232, http://dx.doi.org/10. jwmr.2018.00283.
1111/ics.12036. [85] D.R. Opel, E. Hagstrom, A.K. Pace, K. Sisto, S.A. Hirano-Ali, S. Desai, J. Swan,
[63] B.H. Mahmoud, C.L. Hexsel, I.H. Hamzavi, H.W. Lim, Effects of visible light on Light- emitting diodes: a brief review and clinical experience, J. Clin.
the skin, Photochem. Photobiol. 84 (2) (2008) 450–462, http://dx.doi.org/10. Aesthet. Dermatol. 8 (6) (2015) 36–44.
1111/j.1751-1097.2007.00286.x. [86] H. Serrage, V. Heiskanen, W.M. Palin, P.R. Cooper, M.R. Milward, M. Hadis,
[64] J. D’Orazio, S. Jarrett, A. Amaro-Ortiz, T. Scott, UV radiation and the skin, Int. M.R. Hamblin, Under the spotlight: mechanisms of photobiomodulation
J. Mol. Sci. 14 (6) (2013) 12222–12248, http://dx.doi.org/10.3390/ concentrating on blue and green light, Photochem. Photobiol. Sci. 18 (8)
ijms140612222. (2019) 1877–1909, http://dx.doi.org/10.1039/c9pp00089e.
[65] M. Clement, G. Daniel, M. Trelles, Optimising the design of a broad-band [87] C. Mignon, N.E. Uzunbajakava, B. Raafs, N.V. Botchkareva, D.J. Tobin,
light source for the treatment of skin, J. Cosmet. Laser Ther. 7 (3-4) (2005) Photobiomodulation of human dermal fibroblasts in vitro: decisive role of
177–189, http://dx.doi.org/10.1080/14764170500344575. cell culture conditions and treatment protocols on experimental outcome,
[66] C. Ash, M. Dubec, K. Donne, T. Bashford, Effect of wavelength and beam Sci. Rep. 7 (1) (2017) 2797, http://dx.doi.org/10.1038/s41598-017-02802-0.
width on penetration in light-tissue interaction using computational [88] A. Slominski, J. Wortsman, Neuroendocrinology of the skin, Endocr. Rev. 21
methods, Lasers Med. Sci. 32 (8) (2017) 1909–1918, http://dx.doi.org/10. (5) (2000) 457–487, http://dx.doi.org/10.1210/edrv.21.5.0410.
1007/s10103-017-2317-4. [89] A.T. Slominski, M.A. Zmijewski, C. Skobowiat, B. Zbytek, R.M. Slominski, J.D.
[67] A.J. Ridley, J.R. Whiteside, T.J. McMillan, S.L. Allinson, Cellular and Steketee, Sensing the environment: regulation of local and global
sub-cellular responses to UVA in relation to carcinogenesis, Int. J. Radiat. homeostasis by the skin’s neuroendocrine system, Adv. Anat. Embryol. Cell
Biol. 85 (3) (2009) 177–195, http://dx.doi.org/10.1080/ Biol. 212 (vii) (2012) 1–115, http://dx.doi.org/10.1007/978-3-642-19683-6
09553000902740150. 1.
[68] R.M. Brand, P. Wipf, A. Durham, M.W. Epperly, J.S. Greenberger, L.D. Falo Jr, [90] D. Raj, D.E. Brash, D. Grossman, Keratinocyte apoptosis in epidermal
Targeting mitochondrial oxidative stress to mitigate UV-induced skin development and disease, J. Invest. Dermatol. 126 (2) (2006) 243–257,
damage, Front. Pharmacol. 9 (2018) 920, http://dx.doi.org/10.3389/fphar. http://dx.doi.org/10.1038/sj.jid.5700008.
2018.00920. [91] A. Costanzo, F. Fausti, G. Spallone, F. Moretti, A. Narcisi, E. Botti, Programmed
[69] L.H.F. Mullenders, Solar UV damage to cellular DNA: from mechanisms to cell death in the skin, Int. J. Dev. Biol. 59 (1-3) (2015) 73–78, http://dx.doi.
biological effects, Photochem. Photobiol. Sci. 17 (12) (2018) 1842–1852, org/10.1387/ijdb.150050ac.
http://dx.doi.org/10.1039/c8pp00182k. [92] D. Mohania, S. Chandel, P. Kumar, V. Verma, K. Digvijay, D. Tripathi, K.
[70] C. Skobowiat, A.T. Slominski, UVB activates hypothalamic-pituitary-adrenal Choudhury, S.K. Mitten, D. Shah, Ultraviolet radiations: skin
axis in C57BL/6 mice, J. Invest. Dermatol. 135 (6) (2015) 1638–1648, http:// defense-damage mechanism, Adv. Exp. Med. Biol. 996 (2017) 71–87, http://
dx.doi.org/10.1038/jid.2014.450. dx.doi.org/10.1007/978-3-319-56017-5 7.
[71] I. Albers, E. Zernickel, M. Stern, M. Broja, H.L. Busch, C. Heiss, V. Grotheer, J. [93] P. Dakup, S. Gaddameedhi, Impact of the circadian clock on UV-induced
Windolf, C.V. Suschek, Blue light (lambda=453nm) nitric oxide dependently DNA damage response and photocarcinogenesis, Photochem. Photobiol. 93
induces beta-endorphin production of human skin keratinocytes in-vitro (1) (2017) 296–303, http://dx.doi.org/10.1111/php.12662.
and increases systemic beta-endorphin levels in humans in-vivo, Free Radic. [94] M. Rinnerthaler, J. Bischof, M.K. Streubel, A. Trost, K. Richter, Oxidative
Biol. Med. 145 (2019) 78–86, http://dx.doi.org/10.1016/j.freeradbiomed. stress in aging human skin, Biomolecules 5 (2) (2015) 545–589, http://dx.
2019.09.022. doi.org/10.3390/biom5020545.
[72] M.F. Holick, Ultraviolet B radiation: the vitamin d connection, Adv. Exp. Med. [95] T.G. Polefka, T.A. Meyer, Cutaneous oxidative stress and aging, in: M.A.
Biol. 996 (2017) 137–154, http://dx.doi.org/10.1007/978-3-319-56017-5 12. Farage, K.W. Miller, H.I. Maibach (Eds.), Textbook of Aging Skin., Springer,
[73] J. Liebmann, M. Born, V. Kolb-Bachofen, Blue-light irradiation regulates Berlin Heidelberg, Berlin, Heidelberg, 2016, pp. 651–676, http://dx.doi.org/
proliferation and differentiation in human skin cells, J. Invest. Dermatol. 130 10.1007/978-3-662-47398-6 123.
(1) (2010) 259–269, http://dx.doi.org/10.1038/jid.2009.194. [96] H. Wang, E.Q. van Spyk, M. Geyfman, M.L. Salmans, V. Kumar, A. Ihler, N.
[74] S. Buscone, A.N. Mardaryev, B. Raafs, J.W. Bikker, C. Sticht, N. Gretz, N. Farjo, Ning, J.S. Takahashi, B. Andersen, Time-restricted feeding shifts the skin
N.E. Uzunbajakava, N.V. Botchkareva, A new path in defining light circadian clock and alters UVB-induced DNA damage, Cell Rep. 20 (5) (2017)
parameters for hair growth: discovery and modulation of photoreceptors in 1061–1072, http://dx.doi.org/10.1016/j.celrep.2017.07.022.
human hair follicle, Lasers Surg. Med. 49 (7) (2017) 705–718, http://dx.doi. [97] L.V.M. de Assis, M.N. Moraes, A.M.L. Castrucci, The molecular clock in the
org/10.1002/lsm.22673. skin, its functionality, and how it is disrupted in cutaneous melanoma: a
[75] H.J. Kim, M.S. Choi, I.H. Bae, J.Y. Jung, E.D. Son, T.R. Lee, D.W. Shin, Short new pharmacological target? Cell. Mol. Life Sci. 76 (19) (2019) 3801–3826,
wavelength visible light suppresses innate immunity-related responses by http://dx.doi.org/10.1007/s00018-019-03183-5.
modulating protein S-nitrosylation in keratinocytes, J. Invest. Dermatol. 136 [98] P.S. Welz, V.M. Zinna, A. Symeonidi, K.B. Koronowski, K. Kinouchi, J.G. Smith,
(3) (2016) 727–731, http://dx.doi.org/10.1016/j.jid.2015.12.004. I.M. Guillén, A. Castellanos, S. Furrow, F. Aragón, G. Crainiciuc, N. Prats, J.M.
[76] P.S. Oh, H. Hwang, H.S. Jeong, J. Kwon, H.S. Kim, M. Kim, S. Lim, M.H. Sohn, Caballero, A. Hidalgo, P. Sassone-Corsi, S.A. Benitah, BMAL1-driven tissue
H.J. Jeong, Blue light emitting diode induces apoptosis in lymphoid cells by clocks respond independently to light to maintain homeostasis, Cell 177 (6)
stimulating autophagy, Int. J. Biochem. Cell Biol. 70 (2016) 13–22, http://dx. (2019) 1436–1447, http://dx.doi.org/10.1016/j.cell.2019.05.009.
doi.org/10.1016/j.biocel.2015.11.004. [99] W. Schlosser, T. Schmidt-Kaler, E.F. Milone, W. Schlosser, T. Schmidt-Kaler,
[77] G. Sikka, G.P. Hussmann, D. Pandey, S. Cao, D. Hori, J.T. Park, J. Steppan, J.H. E.F. Milone, Atmospheric effects on starlight and sunlight, in: Challenges
Kim, V. Barodka, A.C. Myers, L. Santhanam, D. Nyhan, M.K. Halushka, R.C. of.Astronomy, Springer, Berlin, 1991, pp. 98–105, http://dx.doi.org/10.1007/
Koehler, S.H. Snyder, L.A. Shimoda, D.E. Berkowitz, Melanopsin mediates 978-1-4612-4434-9 19.
light-dependent relaxation in blood vessels, Proc. Natl. Acad. Sci. U. S. A. 111 [100] M. Bohm, I. Holmer, H. Nilsson, O. Noren, Thermal effect of glazing in
(50) (2014) 17977–17982, http://dx.doi.org/10.1073/pnas.1420258111. driver’s cabs: evaluation of the impact of different types of glazing on the
[78] H.J. Kim, E.D. Son, J.Y. Jung, H. Choi, T.R. Lee, D.W. Shin, Violet light down- thermal comfort, Semant. Sch.- Environ. Sci. (2002),
regulates the expression of specific differentiation markers through https://www.semanticscholar.org/paper/Thermal-effect-of-glazing-in-
rhodopsin in normal human epidermal keratinocytes, PLoS One 8 (9) (2013), driver’s-cabs-%3A-of-of-Bohm-
8, http://dx.doi.org/10.1371/journal.pone.007367, e73678. Holmér/90aaa26b5bc4f41658d020104a95662635619841#citing-papers.
[79] M.H. Catao, R.O. Costa, C.F. Nonaka, R.L. Junior, I.R. Costa, Green LED light has [101] A. Hanafi, T. Gharbi, J.-Y. Cornu, In vivo measurement of lower back
anti-inflammatory effects on burns in rats, Burns 42 (2) (2016) 392–396, deformations with Fourier-transform profilometry, Appl. Opt. 44 (2005)
http://dx.doi.org/10.1016/j.burns.2015.07.003. 2266–2273, http://dx.doi.org/10.1364/AO.44.002266.
[80] T.M.S. Simoes, J.A. Fernandes Neto, T.K.B. de Oliveira, C.F.W. Nonaka, M. [102] R. Anderson, J.A. Parrish, The optics of human skin, J. Invest. Dermatol. 77
Catao, Photobiomodulation of red and green lights in the repair process of (1981) 13–19, http://dx.doi.org/10.1111/1523-1747.ep12479191.
third-degree skin burns, Lasers Med. Sci. 35 (1) (2020) 51–61, http://dx.doi. [103] P. Schroeder, C. Calles, T. Benesova, F. Macaluso, J. Krutmann,
org/10.1007/s10103-019-02776-7, 2020. Photoprotection beyond ultraviolet radiation – effective sun protection has
[81] L. Chen, Z. Xu, M. Jiang, C. Zhang, X. Wang, L. Xiang, Light-emitting diode to include protection against infrared A radiation-induced skin damage,
585nm photomodulation inhibiting melanin synthesis and inducing Skin Pharmacol. Physiol. 23 (2010) 15–17.
autophagy in human melanocytes, J. Dermatol. Sci. 89 (1) (2018) 11–18, [104] Scenihr, Scientific Committee on Emerging and Newly Identified Health
http://dx.doi.org/10.1016/j.jdermsci.2017.10.001. Risks SCENIHR Health Effects of Artificial Light, Report, 2012, http://dx.doi.
[82] Y.J. Kim, H.J. Kim, H.L. Kim, H.J. Kim, H.S. Kim, T.R. Lee, D.W. Shin, Y.R. Seo, A org/10.2772/8624.
protective mechanism of visible red light in normal human dermal [105] V.V. Barun, A.P. Ivanov, A.V. Volotovskaya, Absorption spectra and light
fibroblasts: enhancement of GADD45A-mediated DNA repair activity, J. penetration depth of normal and pathologically altered human skin, J. Appl.
Invest. Dermatol. 137 (2) (2017) 466–474, http://dx.doi.org/10.1016/j.jid. Spectrosc. 74 (2007) 430–439.
2016.07.041. [106] R. Henderson, K. Schulmeister, Laser Safety, CRC Press, Boca Raton, 2004, pp.
474, http://dx.doi.org/10.1201/9781420034042.

15
L.V.M. de Assis, P.N. Tonolli, M.N. Moraes et al. Journal of Photochemistry and Photobiology C: Photochemistry Reviews 47 (2021) 100403

[107] J. Leff, N.I. Krinsky, A mutagenic effect of visible light mediated by importance, Proc. SPIE 1403, Laser Appl. Life Sci. (1991), http://dx.doi.org/
endogenous pigments in Euglena gracilis, Science 158 (3806) (1967) 10.1117/12.57282.
1332–1335, http://dx.doi.org/10.1126/science.158.3806.1332. [130] A.F. Uchoa, P.P. Knox, R. Turchielle, N.K. Seifullina, M.S. Baptista, Singlet
[108] G.P. Pfeifer, Y.-H. You, A. Besaratinia, Mutations induced by ultraviolet light, oxygen generation in the reaction centers of Rhodobacter sphaeroides, Eur.
Mutat. Res. Mol. Mech. Mutagen. 571 (2005) 19–31, http://dx.doi.org/10. Biophys. J. 37 (2008) 843–850, http://dx.doi.org/10.1007/s00249-008-0287-
1016/j.mrfmmm.2004.06.057. y.
[109] G.P. Pfeifer, A. Besaratinia, UV wavelength-dependent DNA damage and [131] M. Rózanowska, J. Wessels, M. Boulton, J.M. Burke, M.A.J. Rodgers, T.G.
human non- melanoma and melanoma skin cancer, Photochem. Photobiol. Truscott, T. Sarna, Blue light-induced singlet oxygen generation by retinal
Sci. 11 (2012) 90–97, http://dx.doi.org/10.1039/C1PP05144J. lipofuscin in non-polar media, Free Radic. Biol. Med. 24 (1998) 1107–1112,
[110] A. Deaton, A. Bird, CpG islands and the regulation of transcription, Genes http://dx.doi.org/10.1016/S0891- 5849(97)00395-X.
Dev. 25 (2011) 1010–1022, http://dx.doi.org/10.1101/gad.2037511.1010. [132] J.L. Spudich, C.S. Yang, K.H. Jung, E.N. Spudich, Retinylidene proteins:
[111] J. Cadet, E. Sage, T. Douki, Ultraviolet radiation-mediated damage to cellular structures and functions from archaea to humans, Annu. Rev. Cell Dev. Biol.
DNA, Mutat. Res. – Fundam. Mol. Mech. Mutagen. 571 (1-2) (2005) 3–17, 16 (2000) 365–392, http://dx.doi.org/10.1146/annurev.cellbio.16.1.365.
http://dx.doi.org/10.1016/j.mrfmmm.2004.09.012. [133] V.J. Kefalov, Rod and cone visual pigments and phototransduction through
[112] J. Li, T. Uchida, T. Todo, T. Kitagawa, Similarities and differences between pharmacological, genetic, and physiological approaches, J. Biol. Chem. 287
cyclobutane pyrimidine dimer photolyase and (6-4) photolyase as revealed (3) (2012) 1635–1641, http://dx.doi.org/10.1074/jbc.R111.303008.
by resonance Raman spectroscopy: electron transfer from the FAD cofactor [134] A. Terakita, T. Nagata, Functional properties of opsins and their contribution
to ultraviolet-damaged DNA, J. Biol. Chem. 281 (35) (2006) 25551–25559, to light- sensing physiology, Zool. Sci. 31 (10) (2014) 653–659, http://dx.doi.
http://dx.doi.org/10.1074/jbc.M604483200. org/10.2108/zs140094.
[113] T.T. Tasso, J.C. Schlothauer, H.C. Junqueira, T.A. Matias, K. Araki, É. Liandra- [135] D. Shen, M. Jiang, W. Hao, L. Tao, M. Salazar, H.K. Fong, A human
Salvador, F.C.T. Antonio, P. Homem-De-Mello, M.S. Baptista, Photobleaching opsin-related gene that encodes a retinaldehyde binding protein,
efficiency parallels the enhancement of membrane damage for Biochemistry 33 (1994) 13117–13125, http://dx.doi.org/10.1021/
porphyrazine photosensitizers, J. Am. Chem. Soc. 141 (2019) 15547–15556, bi00248a022.
http://dx.doi.org/10.1021/jacs.9b05991. [136] M. Koyanagi, A. Terakita, K. Kubokawa, Y. Shichida, Amphioxus homologs of
[114] M.S. Baptista, J. Cadet, P. Di Mascio, A.A. Ghogare, A. Greer, M.R. Hamblin, C. Go- coupled rhodopsin and peropsin having 11-cis- and all-trans-retinals as
Lorente, S.C. Nunez, M.S. Ribeiro, A.H. Thomas, M. Vignoni, T.M. Yoshimura, their chromophores, FEBS Lett. 531 (2002) 525–528, http://dx.doi.org/10.
Type I and type II photosensitized oxidation reactions: guidelines and 1016/s0014-5793(02)03616-5.
mechanistic pathways, Photochem. Photobiol. 93 (2017) 912–919, http://dx. [137] T. Okano, T. Yoshizawa, Y. Fukada, Pinopsin is a chicken pineal
doi.org/10.1111/php.12716. photoreceptive molecule, Nature 372 (1994) 94–97, http://dx.doi.org/10.
[115] I.O.L. Bacellar, M.C. Oliveira, L.S. Dantas, E.B. Costa, H.C. Junqueira, W.K. 1038/372094a0.
Martins, A.M. Durantini, G. Cosa, P. Di Mascio, M. Wainwright, R. Miotto, [138] M. Max, P.J. McKinnon, K.J. Seidenman, R.K. Barrett, M.L. Applebury, J.S.
R.M. Cordeiro, S. Miyamoto, M.S. Baptista, Photosensitized membrane Takahashi, R.F. Margolskee, Pineal opsin: a nonvisual opsin expressed in
permeabilization requires contact- dependent reactions between chick pineal, Science 267 (1995) 1502–1506, http://dx.doi.org/10.1126/
photosensitizer and lipids, J. Am. Chem. Soc. 140 (30) (2018) 9606–9615, science.7878470.
http://dx.doi.org/10.1021/jacs.8b05014. [139] S. Kawamura, S. Yokoyama, Expression of visual and nonvisual opsins in
[116] J. Baier, T. Maisch, M. Maier, E. Engel, M. Landthaler, W. Bäumler, Singlet American chameleon, Vis. Res. 37 (1997) 1867–1871, http://dx.doi.org/10.
oxygen generation by UVA light exposure of endogenous photosensitizers, 1016/S0042-6989(96)00309-4.
Biophys. J. 91 (2006) 1452–1459, http://dx.doi.org/10.1529/biophysj.106. [140] T. Yoshikawa, Y. Yashiro, T. Oishi, K. Kokame, Y. Fukada, Immunoreactivities
082388. to rhodopsin and rod/cone transducin antisera in the retina, pineal complex
[117] G.T. Wondrak, M.K. Jacobson, E.L. Jacobson, Endogenous and deep brain of the bullfrog, Rana catesbeiana, Zool. Sci. 11 (5) (1994)
UVA-photosensitizers: mediators of skin photodamage and novel targets for 675–680.
skin photoprotection, Photochem. Photobiol. Sci. 5 (2006) 215–237, http:// [141] S. Yokoyama, H. Zhang, Cloning and characterization of the pineal
dx.doi.org/10.1039/B504573H. gland-specific opsin gene of marine lamprey (Petromyzon marinus), Gene
[118] Y. Miyachi, Photoaging from an oxidative standpoint, J. Dermatol. Sci. 9 (2) 202 (1-2) (1997) 89–93, http://dx.doi.org/10.1016/s0378-1119(97)00458-
(1995) 79–86, http://dx.doi.org/10.1016/0923-1811(94)00363-J. 7.
[119] A.A. Gorman, I. Hamblett, M.A.J. Rodgers, Ergosterol (provitamin D2) triplet [142] I. Provencio, G. Jiang, W.J. De Grip, W.P. Hayes, M.D. Rollag, Melanopsin: an
state: an efficient sensitizer of singlet oxygen, O2(g), formation, opsin in melanophores, brain, and eye, Proc. Natl. Acad. Sci. U. S. A. 95
Photochem. Photobiol. 45 (2) (1987) 215–221, http://dx.doi.org/10.1111/j. (1998) 340–345, http://dx.doi.org/10.1073/pnas.95.1.340.
1751-1097.1987.tb05366.x. [143] I. Provencio, I.R. Rodriguez, G. Jiang, W.P. Hayes, E.F. Moreira, M.D. Rollag, A
[120] A.A. Gorman, I. Hamblett, C. Lambert, A.L. Prescott, A pulse radiolysis and novel human opsin in the inner retina, J. Neurosci. 20 (2000) 600–605,
pulsed laser study of the vitamin D3 triplet state: lifetime, relaxation and http://dx.doi.org/10.1523/JNEUROSCI.20-02-00600.2000.
nonvertical excitation, Photochem. Photobiol. 51 (1) (1990) 29–35, http:// [144] S. Panda, I. Provencio, D.C. Tu, S.S. Pires, M.D. Rollag, A.M. Castrucci, M.T.
dx.doi.org/10.1111/j.1751-1097.1990.tb01680.x. Pletcher, T.K. Sato, T. Wiltshire, M. Andahazy, S.A. Kay, R.N. Van Gelder, J.B.
[121] E. Oliveros, F. Besançon, M. Boneva, B. Kräutler, A.M. Braun, Singlet oxygen Hogenesch, Melanopsin is required for non-image-forming photic responses
(1g) sensitization and quenching by vitamin B12 derivatives, J. in blind mice, Science 301 (5632) (2003) 525–527, http://dx.doi.org/10.
Photochem. Photobiol. B, Biol. 29 (1) (1995) 37–44, http://dx.doi.org/10. 1126/science.1086179.
1016/1011-1344(95)90249-X. [145] Y. Miyashita, T. Moriya, T. Kubota, K. Yamada, K. Asami, Expression of opsin
[122] A. Feis, B. Wegewijs, W. Gärtner, S.E. Braslavsky, Role of the triplet state in molecule in cultured murine melanocyte, J. Investig. Dermatol. Symp. Proc.
retinal photoisomerization as studied by laser-induced optoacoustic 6 (2001) 54–57, http://dx.doi.org/10.1046/j.0022-202x.2001.00018.x.
spectroscopy, J. Phys. Chem. B 101 (38) (1997) 7620–7627, http://dx.doi.org/ [146] G.J. Lopes, C.C. Gois, L.H. Lima, A.M. Castrucci, Modulation of rhodopsin gene
10.1021/jp970879d. expression and signaling mechanisms evoked by endothelins in goldfish and
[123] K. Bhattacharyya, P.K. Das, Quantitative aspects of all-trans-retinol singlet murine pigment cell lines, Braz. J. Med. Biol. Res. 43 (2010) 828–836, http://
and triplet quenching by oxygen, Chem. Phys. Lett. 116 (4) (1985) 326–332, dx.doi.org/10.1590/s0100- 879x2010007500087.
http://dx.doi.org/10.1016/0009- 2614(85)80178-0. [147] J. Bellingham, D. Whitmore, A.R. Philp, D.J. Wells, R.G. Foster, Zebrafish
[124] A.V. Silva, A. López-Sánchez, H.C. Junqueira, L. Rivas, M.S. Baptista, G. melanopsin: isolation, tissue localisation and phylogenetic position, Brain
Orellana, Riboflavin derivatives for enhanced photodynamic activity against Res. Mol. Brain Res. 107 (2) (2002) 128–136, http://dx.doi.org/10.1016/
Leishmania parasites, Tetrahedron 71 (3) (2015) 457–462, http://dx.doi.org/ s0169-328x(02)00454-0.
10.1016/j.tet.2014.11.072. [148] N. Oshima, Direct reception of light by chromatophores of lower
[125] E. Consiglieri, Q. Xu, M. Bregnhøj, M. Westberg, P.R. Ogilby, A. Losi, Single vertebrates, Pigment Cell Res. 14 (5) (2001) 312–319, http://dx.doi.org/10.
mutation in a novel bacterial LOV protein yields a singlet oxygen generator, 1034/j.1600-0749.2001.140502.x.
Photochem. Photobiol. Sci. 18 (2019) 2657–2660, http://dx.doi.org/10.1039/ [149] M. Tsutsumi, K. Ikeyama, S. Denda, J. Nakanishi, S. Fuziwara, H. Aoki, M.
c9pp00328b. Denda, Expressions of rod and cone photoreceptor-like proteins in human
[126] A.H. Thomas, C. Lorente, A.L. Capparelli, C.G. Martínez, A.M. Braun, E. epidermis, Exp. Dermatol. 18 (6) (2009) 567–570, http://dx.doi.org/10.1111/
Oliveros, Singlet oxygen (1g) production by pterin derivatives in aqueous j.1600-0625.2009.00851.x.
solutions, Photochem. Photobiol. Sci. 2 (2003) 245–250, http://dx.doi.org/ [150] N.L. Wicks, J.W. Chan, J.A. Najera, J.M. Ciriello, E. Oancea, UVA
10.1039/b209993d. phototransduction drives early melanin synthesis in human melanocytes,
[127] S.M. Bishop, M. Malone, D. Phillips, A.W. Parker, M.C.R. Symons, Singlet Curr. Biol. 21 (22) (2011) 1906–1911, http://dx.doi.org/10.1016/j.cub.2011.
oxygen sensitisation by excited state DNA, J. Chem. Soc. Chem. Commun. 09.047.
1994 (1994) 871–872, http://dx.doi.org/10.1039/C39940000871. [151] M. Denda, S. Fuziwara, Visible radiation affects epidermal permeability
[128] K.K. Chin, C.C. Trevithick-Sutton, J. McCallum, S. Jockusch, N.J. Turro, J.C. barrier recovery: selective effects of red and blue light, J. Invest. Dermatol.
Scaiano, C.S. Foote, M.A. Garcia-Garibay, Quantitative determination of 28 (2008) 1335–1336, http://dx.doi.org/10.1038/sj.jid.5701168.
singlet oxygen generated by excited state aromatic amino acids, proteins, [152] D. Kojima, S. Mori, M. Torii, A. Wada, R. Morishita, Y. Fukada, UV-sensitive
and immunoglobulins, J. Am. Chem. Soc. 52 (14) (2008) 7671–7679, http:// photoreceptor protein OPN5 in humans and mice, PLoS One 6 (10) (2011),
dx.doi.org/10.1021/ja800926v. http://dx.doi.org/10.1371/journal.pone.0026388, e26388.
[129] S.Y. Egorov, A.A. Krasnovsky Jr, Laser-induced luminescence of singlet [153] L.V.M. de Assis, M.N. Moraes, S.S. Cruz-Machado, A.M.L. Castrucci, The effect
molecular oxygen: generation by drugs and pigments of biological of white light on normal and malignant murine melanocytes: A link between

16
L.V.M. de Assis, P.N. Tonolli, M.N. Moraes et al. Journal of Photochemistry and Photobiology C: Photochemistry Reviews 47 (2021) 100403

opsins, clock genes, and melanogenesis, Biochim. Biophys. Acta 1863 (6 Pt [176] W.L. Shen, Y. Kwon, A.A. Adegbola, J. Luo, A. Chess, C. Montell, Function of
A) (2016) 1119–1133, http://dx.doi.org/10.1016/j.bbamcr.2016.03.001. rhodopsin in temperature discrimination in Drosophila, Science 331 (6022)
[154] P.P. Toh, M. Bigliardi-Qi, A.M.Y. Yap, G. Sriram, O. Stelmashenko, P. Bigliardi, (2011) 1333–1336, http://dx.doi.org/10.1126/science.1198904.
Expression of peropsin in human skin is related to phototransduction of [177] H.B. Barlow, The thermal limit to seeing, Nature 334 (1988) 296–297, http://
violet light in keratinocytes, Exp. Dermatol. 25 (12) (2016) 1002–1005, dx.doi.org/10.1038/334296a0.
http://dx.doi.org/10.1111/exd.13226. [178] R.B. Barlow, R.R. Birge, E. Kaplan, J.R. Tallent, On the molecular origin of
[155] Y. Lan, Y. Wang, H. Lu, Opsin 3 is a key regulator of ultraviolet A-induced photoreceptor noise, Nature 366 (1993) 64–66, http://dx.doi.org/10.1038/
photoaging in human dermal fibroblast cells, Br. J. Dermatol. 182 (5) (2019) 366064a0, 1993.
1228–1244, http://dx.doi.org/10.1111/bjd.18410. [179] Y. Guo, H.P. Hendrickson, P.E. Videla, Y.-N. Chen, J. Ho, S. Sekharan, V.S.
[156] J. Kusumoto, M. Takeo, K. Hashikawa, T. Komori, T. Tsuji, H. Terashi, S. Batista, J.C. Tully, E.C.Y. Yan, Probing the remarkable thermal kinetics of
Sakakibara, OPN4 belongs to the photosensitive system of the human skin, visual rhodopsin with E181Q and S186A mutants, J. Chem. Phys. 146 (2017)
Genes Cells 25 (3) (2020) 215–225, http://dx.doi.org/10.1111/gtc.12751. 215104, http://dx.doi.org/10.1063/1.4984818.
[157] G. Hinterhuber, K. Cauza, K. Brugger, R. Dingelmaier-Hovorka, R. Horvat, K. [180] T. Sokabe, H.C. Chen, J. Luo, C. Montell, A switch in thermal preference in
Wolff, D. Foedinger, RPE65 of retinal pigment epithelium, a putative Drosophila larvae depends on multiple rhodopsins, Cell Rep. 17 (2) (2016)
receptor molecule for plasma retinol-binding protein, is expressed in 336–344, http://dx.doi.org/10.1016/j.celrep.2016.09.028.
human keratinocytes, J. Invest. Dermatol. 122 (2) (2004) 406–413, http://dx. [181] S. Pérez-Cerezales, S. Boryshpolets, O. Afanzar, A. Brandis, R. Nevo, V. Kiss,
doi.org/10.1046/j.0022-202X.2004.22216.x. M. Eisenbach, Involvement of opsins in mammalian sperm thermotaxis, Sci.
[158] Y. Shichida, T. Matsuyama, Evolution of opsins and phototransduction, Rep. 5 (2015) 16146, http://dx.doi.org/10.1038/srep16146.
Philos. Trans. R. Soc. B Biol. Sci. 364 (1531) (2009) 2881–2895, http://dx.doi. [182] D. Roy, K. Levi, V. Kiss, R. Nevo, M. Eisenbach, Rhodopsin and melanopsin
org/10.1098/rstb.2009.0051. coexist in mammalian sperm cell and activate different signaling pathways
[159] R. Birge, Photophysics and molecular electronic applications of the for thermotaxis, Sci. Rep. 10 (1) (2020) 112, http://dx.doi.org/10.1038/
rhodopsins, Annu. Rev. Phys. Chem. 41 (1990) 683–733, doi: s41598-019-56846-5.
10.1146/annurev.pc.41.10 0190.003343. [183] M.N. Moraes, L.V.M. de Assis, K.K. Magalhães-Marques, M.O. Poletini,
[160] D. Oesterhelt, W. Stoeckenius, Functions of a new photoreceptor membrane, L.H.R.G. de Lima, A.M.L. Castrucci, Melanopsin, a canonical light receptor,
Proc. Natl. Acad. Sci. U. S. A. 70 (10) (1973) 2853–2857, http://dx.doi.org/10. mediates thermal activation of clock genes, Sci. Rep. 7 (1) (2017) 13977,
1073/pnas.70.10.2853. http://dx.doi.org/10.1038/s41598-017-13939-3.
[161] K. Inoue, T. Tsukamoto, Y. Sudo, Molecular and evolutionary aspects of [184] N.W. Bellono, J.A. Najera, E. Oancea, UV light activates a Galphaq/11-coupled
microbial sensory rhodopsins, Biochim. Biophys. Acta 1837 (5) (2014) phototransduction pathway in human melanocytes, J. Gen. Physiol. 143
562–577, http://dx.doi.org/10.1016/j.bbabio.2013.05.005. (2014) 203–214, http://dx.doi.org/10.1085/jgp.201311094.
[162] M. Kurihara, Y. Sudo, Microbial rhodopsins: wide distribution, rich diversity [185] T. Sugiyama, H. Suzuki, T. Takahashi, Light-induced rapid Ca2+ response and
and great potential, Biophys. Physicobiol. 12 (2015) 121–129, http://dx.doi. MAPK phosphorylation in the cells heterologously expressing human OPN5,
org/10.2142/biophysico.12.0 121. Sci. Rep. 4 (2014) 5352, http://dx.doi.org/10.1038/srep05352.
[163] A. Cooper, Energy uptake in the first step of visual excitation, Nature 282 [186] L.V.M. de Assis, M.N. Moraes, A.M.L. Castrucci, Heat shock antagonizes UVA-
(5738) (1979) 531–533, http://dx.doi.org/10.1038/282531a0. induced responses in murine melanocytes and melanoma cells: an
[164] E.A. Zhukovsky, D.D. Oprian, Effect of carboxylic acid side chains on the unexpected interaction, Photochem. Photobiol. Sci. 16 (2017) 633–648,
absorption maximum of visual pigments, Science 246 (4932) (1989) http://dx.doi.org/10.1039/c6pp00330c.
928–930, http://dx.doi.org/10.1126/science.2573154. [187] Q.-M. Hu, W.-J. Yi, M.-Y. Su, S. Jiang, S.-Z. Xu, T.-C. Lei, Induction of retinal-
[165] Y. Shichida, H. Imai, Visual pigment: G-protein-coupled receptor for light dependent calcium influx in human melanocytes by UVA or UVB radiation
signals, Cell. Mol. Life Sci. 54 (12) (1998) 1299–1315, http://dx.doi.org/10. contributes to the stimulation of melanosome transfer, Cell Prolif. 50 (2017),
1007/s000180050256. http://dx.doi.org/10.1111/cpr.12372, e12372.
[166] J.E. Kim, M.J. Tauber, R.A. Mathies, Wavelength dependent cis-trans [188] I. Castellano-Pellicena, N.E. Uzunbajakava, C. Mignon, B. Raafs, V.A.
isomerization in vision, Biochemistry 40 (46) (2001) 13774–13778, http:// Botchkarev, M.J. Thornton, Does blue light restore human epidermal barrier
dx.doi.org/10.1021/bi0116137. function via activation of opsin during cutaneous wound healing? Lasers
[167] R.W. Schoenlein, L.A. Peteanu, R.A. Mathies, C.V. Shank, The first step in Surg, Med. 51 (4) (2019) 370–382, http://dx.doi.org/10.1002/lsm.23015.
vision: femtosecond isomerization of rhodopsin, Science 254 (5030) (1991) [189] Y. Lan, Y. Wang, H. Lu, Opsin 3 is a key regulator of ultraviolet A-induced
412–415, http://dx.doi.org/10.1126/science.1925597. photoaging in human dermal fibroblast cells, Br. J. Dermatol. 182 (5) (2019)
[168] D. Polli, P. Altoè, O. Weingart, K.M. Spillane, C. Manzoni, D. Brida, G. 1228–1244, http://dx.doi.org/10.1111/bjd.18410.
Tomasello, G. Orlandi, P. Kukura, R.A. Mathies, M. Garavelli, G. Cerullo, [190] Y. Wang, Y. Lan, H. Lu, Opsin3 downregulation induces apoptosis of human
Conical intersection dynamics of the primary photoisomerization event in epidermal 509 melanocytes via mitochondrial pathway, Photochem.
vision, Nature 467 (2010) 440–443, http://dx.doi.org/10.1038/nature09346. Photobiol. 96 (2020) 83–93, http://dx.doi.org/10.1111/php.13178.
[169] G. Moiseyev, Y. Chen, Y. Takahashi, B.X. Wu, J.X. Ma, RPE65 is the [191] L.V.M. de Assis, D. Mendes, M.M. Silva, G.S. Kinker, I. Pereira-Lima, M.N.
isomerohydrolase in the retinoid visual cycle, Proc. Natl. Acad. Sci. U. S. A. Moraes, C.F.M. Menck, A.M.L. Castrucci, Melanopsin mediates
102 (35) (2005) 12413–12418, http://dx.doi.org/10.1073/pnas.0503460102. UVA-dependent modulation of proliferation, pigmentation, apoptosis, and
[170] T. Matsuyama, T. Yamashita, Y. Imamoto, Y. Shichida, Photochemical molecular clock in normal and malignant melanocytes, BBA – Mol. Cell. Res.
properties of mammalian melanopsin, Biochemistry 51 (27) (2012) 1867 (2020) 118789, http://dx.doi.org/10.1016/j.bbamcr.2020.118789.
5454–5462, http://dx.doi.org/10.1021/bi3004999. [192] H. Dumbuya, S.Y. Hafez, E. Oancea, Cross talk between calcium and ROS
[171] M. Koyanagi, K. Kubokawa, H. Tsukamoto, Y. Shichida, A. Terakita, regulates the UVA-induced melanin response in human melanocytes, FASEB
Cephalochordate melanopsin: evolutionary linkage between invertebrate J. 34 (9) (2020) 11605–11623, http://dx.doi.org/10.1096/fj.201903024R.
visual cells and vertebrate photosensitive retinal ganglion cells, Curr. Biol. [193] E.D. Buhr, S. Vemaraju, N. Diaz, R.A. Lang, R.N. Van Gelder, Neuropsin (OPN5)
15 (11) (2005) 1065–1069, http://dx.doi.org/10.1016/j.cub.2005.04.063. mediates local light-dependent induction of circadian clock genes and
[172] T. Sugihara, T. Nagata, B. Mason, M. Koyanagi, A. Terakita, Absorption circadian photoentrainment in exposed murine skin, Curr. Biol. 29 (20)
characteristics of vertebrate non-visual opsin, Opn3, PLoS One 11 (8) (2016), (2019) 3478–3487, http://dx.doi.org/10.1016/j.cub.2019.08.063.
http://dx.doi.org/10.1371/journal.pone.0161215, e0161215. [194] S.M. Fan, Y.T. Chang, C.L. Chen, W.H. Wang, M.K. Pan, W.P. Chen, W.Y. Huang,
[173] S.G. Solomon, P. Lennie, The machinery of colour vision, Nat. Rev. Neurosci. Z. Xu, H.E. Huang, T. Chen, M.V. Plikus, S.K. Chen, S.J. Lin, External light
8 (4) (2007) 276–286, http://dx.doi.org/10.1038/nrn2094. activates hair follicle stem cells through eyes via an ipRGC-SCN-sympathetic
[174] N.W. Bellono, E. Oancea, UV light phototransduction depolarizes human neural pathway, Proc. Natl. Acad. Sci. U. S. A. 115 (29) (2018) E6880–e6889,
melanocytes, Channels 7 (2013) 243–248, http://dx.doi.org/10.4161/chan. http://dx.doi.org/10.1073/pnas.1719548115.
25322. [195] Y. Leung, D.P. Thakur, A.S. Gurav, S.H. Kim, A. Di Pizio, M.Y. Niv, C. Montell,
[175] N.W. Bellono, L.G. Kammel, A.L. Zimmerman, E. Oancea, UV light Functions of opsins in Drosophila taste, Curr. Biol. 30 (8) (2020) 1367–1379,
phototransduction activates transient receptor potential A1 ion channels in http://dx.doi.org/10.1016/j.cub.2020.01.068.
human melanocytes, Proc. Natl. Acad. Sci. U. S. A. 110 (2013) 2383–2388, [196] M. Wehling, Rapid actions of aldosterone revisited: receptors in the
http://dx.doi.org/10.1073/pnas.1215555110. limelight, J. Steroid Biochem. Mol. Biol. 176 (2018) 94–98, http://dx.doi.org/
10.1016/j.jsbmb.2017.01.016.

17

You might also like