You are on page 1of 15

Accepted: 7 November 2017

DOI: 10.1111/jocd.12518

REVIEW ARTICLE

Atmospheric skin aging—Contributors and inhibitors

David McDaniel MD, FAAD1,2,3,4 | Patricia Farris MD, FAAD5,6 |


Giuseppe Valacchi PhD7,8

1
McDaniel Laser and Cosmetic Center and
McDaniel Institute of Anti-Aging Research, Summary
Virginia Beach, VA, USA Cutaneous aging is a complex biological process consisting of 2 elements: intrinsic
2
Hampton University Skin of Color
aging, which is primarily determined by genetics, and extrinsic aging, which is largely
Research Institute, Hampton, VA, USA
3
School of Science, Hampton University, caused by atmospheric factors, such as exposure to sunlight and air pollution, and
Hampton, VA, USA lifestyle choices, such as diet and smoking. The role of the solar spectrum, com-
4
Department of Biological Sciences, Old
prised of ultraviolet light, specifically UVB (290-320 nm) and UVA (320-400) in
Dominion University, Norfolk, VA, USA
5
Department of Dermatology at Tulane
causing skin damage, including skin cancers, has been well documented. In recent
University, New Orleans, LA, USA years, the contribution of visible light (400-700 nm) and infrared radiation (above
6
Old Metairie Dermatology, Metairie, LA, 800 nm) in causing skin damage, similar to the photodamage caused by UV light, is
USA
7 also being elucidated. In addition, other atmospheric factors such as air pollution
Plants for Human Health Institute,
Department of Animal Sciences, NC State (smog, ozone, particulate matter, etc.) have been implicated in premature skin aging.
University, Kannapolis, NC, USA
8
The skin damage caused by environmental exposure is largely attributable to a com-
Department of Life Sciences and
Biotechnology, University of Ferrara, plex cascade of reactions inside the skin initiated by the generation of reactive oxy-
Ferrara, Italy gen species (ROS), which causes oxidative damage to cellular components such as
Correspondence proteins, lipids, and nucleic acids. These damaged skin cells initiate inflammatory
David McDaniel, McDaniel Laser and responses leading to the eventual damage manifested in chronically exposed skin.
Cosmetic Center and McDaniel Institute of
Anti-Aging Research, Virginia Beach, VA, Novel therapeutic strategies to combat ROS species generation are being developed
USA. to prevent the skin damage caused by atmospheric factors. In addition to protecting
Email: research@drmcdaniel.com
skin from solar radiation using sunscreens, other approaches using topically applied
Funding information ingredients, particularly antioxidants that penetrate the skin and protect the skin
SkinCeuticals, NY
from within, have also been well documented. This review summarizes current
knowledge of atmospheric aggressors, including UVA, UVB, visible light, infrared
radiation (IR), and ozone on skin damage, and proposes new avenues for future
research in the prevention and treatment of premature skin aging caused by such
atmospheric factors. New therapeutic modalities currently being developed are also
discussed.

KEYWORDS
antioxidants, atmospheric skin aging, oxidative stress, ozone, solar radiation

1 | INTRODUCTION exposures to which an individual is subjected from conception to


death. It includes both external and internal factors as well as the
Solar exposure coupled with living in an oxygen-rich atmosphere can human body’s response to these factors.2 Photodamage caused by
cause unwanted and deleterious stress on the skin.1 It has been pre- sunlight can lead to wrinkling, scaling, dryness, and mottled pigment
viously described in the literature that aging entails the totality of abnormalities (hypo- and hyperpigmentation). The extreme

124 | © 2018 Wiley Periodicals, Inc. wileyonlinelibrary.com/journal/jocd J Cosmet Dermatol. 2018;17:124–137.


MCDANIEL ET AL. | 125

consequence of photodamage is skin cancer. The UV portion of sun- inflammation. Near-infrared radiation (IR-A) acts via similar mecha-
light is absorbed by chromophores present in the skin, resulting in nisms as UV, inducing skin damage related to UV.7 In addition to
the generation of reactive oxygen species (ROS) within the skin. solar radiation, air pollution has also been extensively studied for its
These ROS cause damaging oxidation of DNA, including oxidation of effect on human skin. Recent studies have shed light on the complex
nucleic acids, proteins, lipids, and other cellular organelles such as interaction of intrinsic aging that is accelerated by these extrinsic
mitochondria, resulting in photodamage and visible tissue aging.3 atmospheric factors. This review summarizes our current knowledge
Many chromophores (chemicals capable of absorbing UV radia- on solar radiation (UV, visible, IR), as well as air pollution (with an
tion and generating damaging reactive oxygen species) exist within emphasis on ozone) and their effect on skin. We also discuss current
the skin.4 DNA absorbs UVB light, inducing changes between adja- and new therapeutic approaches to preventing and treating atmo-
cent pyrimidine bases. These damaged DNA molecules can stimulate spheric skin aging, including the use of novel topical antioxidants
repair mechanisms that are known to cause inflammatory responses and other agents.
within the skin. Other chromophores within the skin absorb photons
from UV light and generate singlet oxygen. Singlet oxygen, a highly
reactive oxygen species, can, in turn, generate other ROS, and in 2 | PART 1: UV AND SKIN AGING—ROLE
combination, attack cellular components such as cell membranes, cel- OF UVB AND UVA
lular proteins, lipids, and nuclear and mitochondrial DNA.4
Skin is also equipped with protective antioxidant systems, includ- The solar spectrum reaching the surface of the earth is divided into
ing chemical and enzymatic antioxidants. Chemical antioxidants can 3 main segments based on wavelength: UVC (100-290 nm), UVB
be water-soluble (vitamin C, glutathione, lipoic acid, uric acid etc.), or (290-320 nm), and UVA (320-400 nm) (Figure 1). UVA can further
lipid-soluble (vitamin E and other tocopherol forms, ubiquinone, car- be divided into short length UVA2 (320-340) or long length UVA1
otenoids etc.). Enzymatic antioxidants include superoxide dismutase, rays (340-400 nm). The most energetic and highly toxic of the UV
catalase, glutathione peroxidase, and glutathione transferase. These spectrum, UVC, is filtered by atmospheric ozone and moisture, and
antioxidants protect the skin from ROS and free radicals; however, consequently does not reach the earth’s surface. Generally, UVA
with age and environmental stress, such as UV and ozone exposure, represents up to 95% of total UV reaching the earth, with UVB
the levels of these antioxidants are depleted, making the skin more accounting for 4-5%. On a mid-summer day, twenty times more
vulnerable to damage.5,6 A variety of antioxidants can be included in UVA reaches the earth’s surface than UVB8 (Figure 2). UVA is less
topical products to “boost” the endogenous antioxidant systems energetic than UVB, but exhibits higher penetration properties,
within the skin. These include common vitamins already found in reaching the lower dermis and leading to deeper cutaneous damage.
skin, activators of antioxidant enzyme systems present within the The contribution of UVA to human photocarcinogenesis,9 skin pho-
skin, or plant-derived compounds that have strong antioxidant prop- toaging,10 and immunesuppression11 has been well established. Both
4
erties. UVA and UVB have acute and chronic effects on human skin12
Recent research has demonstrated that skin is a target for addi- (Table 1 summarizes the types of solar radiation and their effects on
tional spectra of solar radiation outside the UV bandwidth. For skin).
example, visible light can cause changes in skin similar to that of It has been established that approximately 50% of UV-induced
UVA including the induction of skin pigmentation and skin photodamage is caused by the generation of free radicals, while

FIGURE 1 Solar spectrum


126 | MCDANIEL ET AL.

Figure 3 shows the complex interaction between UV radiation


and skin epidermal and dermal cells. Following sun exposure, activa-
tion of inflammatory reactions within the skin and other cell types
such as monocytes, macrophages, lymphocytes, and vascular
endothelial cells occurs due to upregulation of nuclear factor NF-kB
pathway.19 This signaling pathway causes epidermal keratinocytes to
release primary pro-inflammatory cytokines: tumor necrosis factor
alpha (TNF-a) and interleukin 1 (IL-1).20 Other secondary inflamma-
tory cytokines IL-3, IL-6, IL-8, IL-7, and IL-10 are also released fol-
lowing the initial inflammatory response. UV exposure also activates
the cyclooxygenase and lipoxygenase enzyme systems (COX-2 and
LOX) increasing the production of further pro-inflammatory prosta-
glandins and leukotrienes. Additional factors such as GM-CSF
released by the skin keratinocytes can cause immunosuppression.21
FIGURE 2 Different solar radiations and their penetration into A complex cascade of reactions initiated by the primary inflamma-
skin tory mediators ultimately stimulates factors involved in macrophage
activation.
Photoaging results largely from the effects of UVA on the dermal
direct cellular injury accounts for the remainder of the damage.13 matrix. ROS upregulate transcription factor activator protein 1 (AP-
UV-induced cellular responses occur due to oxidative processes initi- 1), increasing the production of matrix metalloproteinases (MMPs),
ated by cellular photosensitization. After UV absorption by cellular the enzymes that breakdown collagen.19 MMPs such as interstitial
chromophores (DNA, nucleic acids, urocanic acid, aromatic amino collagenase (MMP-1), stromelysin (MMP-3), and gelatinase (MMP-9)
acids tryptophan and tyrosine, and other UV-absorbing endogenous are produced, and keratinocytes, macrophages, and leukocytes lead
molecules such as NADH, quinones, flavins, and porphyrins), singlet to the destruction of dermal matrix proteins. Damage to collagen,
14,15
oxygen, hydrogen peroxide, or hydroxyl radicals are generated. elastin, and glycosaminoglycans contributes to the photoaged pheno-
Most of these endogenous chromophores absorb in the UVB range. type, and causes solar elastosis and wrinkling. Furthermore, AP-1
Although UVA exposure results in more oxidation than UVB, cuta- inhibits collagen production in the dermis by inhibiting transforming
neous UVA chromophores are largely unknown, except for trans- growth factor beta (TGF-ẞ) and reducing gene expression of type I
14
urocanic acid. and III procollagen. Thus, chronic UV exposure results in a net loss
The reactive oxygen species generated by UV can cause damage in dermal collagen due to both degradation and attenuation of pro-
to cellular proteins, lipids, nucleic acids, cell membranes, and cell duction.22
organelles such as mitochondria. UV and infrared (IR-A) exposure Acute exposure to UVB causes sunburn that peaks 6-24 hours
cause disturbance in the electron transport chain resulting in lower after exposure. The redness and swelling associated with sunburn
energy production and decreased mitochondrial function in human are due to the release of pro-inflammatory mediators and vasodila-
fibroblasts.16 tion of dermal blood vessels. UVA and UVB differ in their pigment
UVB is directly absorbed by DNA in epidermal cells causing darkening response and mechanism of action. The short-term con-
the production of characteristic DNA mutations including cyclobu- sequence of UVA exposure is immediate pigment darkening (IPD),
tane pyrimidine dimers and pyrimidine cross-linked dimers.17 The an immediate gray-brown discoloration/pigmentation resulting from
damaged DNA products, particularly pyrimidine dimers, also acti- photooxidation of melanin and its redistribution within the ker-
vate skin melanogenesis, thereby providing protection to skin from atinocytes.17 In contrast, delayed tanning response (DT), caused by
further UV damage.17 The damaged DNA causes a block in the UVB, is due to melanogenesis activation. IPD fades rapidly, but
production of RNA and activation of p53 proteins inducing apop- after sufficient UVA doses (>10 J/cm²), it is followed by a residual
tosis of keratinocytes in the epidermis, leading to the formation pigmentation known as persistent pigment darkening (PPD).23 This
of “sunburn” cells in the epidermis. Prolonged irradiation can sup- is a significant problem for pigmented skin populations, because of
press the p53-mediated apoptosis process resulting in the accumu- their high susceptibility to hyperpigmented lesions.24,25 Most stud-
lation of damaged cells and the initiation of mutagenesis and ies on the damaging effects of UV have been studied in Caucasian
photocarcinogenesis.3 UVA renders DNA damage primarily through skin types; the molecular impact of UV exposure in darker skin
oxidation of guanine to 8-hydroxyguanine resulting in 8-oxogua- phototypes is only beginning to gain attention in recent years.26
nine (8oG). Using RT-PCR methods, it has been demonstrated that the UVA1-
Although the skin possesses an elaborate antioxidant system (de- induced genetic changes in oxidative stress response, inflammation,
scribed in more detail in Section 6) to deal with the UV-induced extracellular remodeling, and cancer development are modulated in
oxidative stress, chronic exposure can overcome the endogenous the same manner in Caucasian, Indian, and African American skin
antioxidant capacity of the skin leading to further damage.18 types.26
MCDANIEL ET AL. | 127

T A B L E 1 Summary of solar radiation and its effects on skin


Effects UVC UVB UVA Visible IR
Wave lengths (nm) 200-290 290-320 UVA-2 320-340 400-700 IR-A = 700-1400
UVA-1 = 340-400 IR-B = 1400-3000
IR-C = 3000-10 000
Energy Very High High Medium Medium-low Low
Amount reaching None. Atmospheric 4-5% of total solar 90% of total UV 40% of total solar Almost half of solar
surface of earth Ozone and moisture UV radiation. radiation reaching radiation reaching energy reaching earth
absorbs it Highest in summer earth is UVA surface of earth surface is IR
and depends on
time of day
Skin Penetration Does not penetrate Epidermal layers Dermis and Down to dermis and Deeper layers of skin
skin subdermis below
Time line for effects None (from solar Immediate within Chronic effects Chronic effects Immediate burning
on skin (Acute vs. exposure) hours sensation with IR-B
chronic) and IR-C
Damage to skin No damage in vivo; Causes skin burning Chronic exposure Damaging effects Oxidative stress on
Highly toxic to cells and erythema and causes dermal similar to UVA skin, rises skin
and kills organisms chronic exposure matrix damage, and temperature,
when exposed can cause eventually “ solar collagenase induction,
in vitro carcinogenesis elastosis” and skin and matrix degradation
cancers
Skin pigmentation None Activates skin Immediate pigment Increased skin IR-A can cause skin
melanogenesis and darkening (IPD) and darkening in darkening in darker
causes skin persistent pigment pigmented skins. skin types
darkening in all skin darkening (PPD), IPD response for IR-
types within days of especially in darker A. No significant
exposure (delayed skin types effects on lighter
tanning response) skin types (I, II)
Mechanism of Epidermal DNA Oxidative stress Oxidative stress, Oxidative stress,
action on skin damage, depletion induction; depletion actinic dermatitis, activation of matrix
of AOX in of AOX in dermis, phototoxic, and degradation in lighter
epidermis, activation MMP activation, photoallergic skin types
of skin inflammatory collagen reactions in skin
reactions, degradation,
photocarcinogenesis inflammatory cell
influx from blood
vessels.
Beneficial effects on Vitamin D synthesis Certain UVA has Therapeutic benefits Used therapeutically for
skin been used to treat for blue light and certain skin disorders
skin wrinkles (blue pulsating yellow
light laser) light for collagen
stimulation
Traditional Sunscreens. Most Certain new broad- Clothing, umbrella Clothing, umbrella and
protective commonly used spectrum and moving out of moving out of direct
measures sunscreens protect sunscreens provide direct sunlight sunlight
skin from UVB partial protection
New Therapeutic Novel sunscreens Novel topical and Topical and systemic Topical and systemic
and protective with antioxidant systemic antioxidants to antioxidants to
measures properties antioxidants to prevent ROS prevent ROS
prevent ROS generation within generation within skin
generation within skin
skin

As oxidative stress and the release of ROS molecules are a common ROS within the skin. Topical application of antioxidants in combination
initial step in the damage caused by UVA, and to a lesser extent UVB, with sunscreens has been used to effectively protect skin from UV
strategies have been developed to protect skin from generation of the damage. This strategy is discussed in detail in part VI of this review.
128 | MCDANIEL ET AL.

FIGURE 3 Skin inflammatory response


to UVR

3 | PART II VISIBLE LIGHT AND SKIN Studies have also shown that a single exposure to visible light in-
AGING duced very little pigmentation, whereas multiple exposures
with visible light resulted in darker and sustained pigmentation.30
Visible light (VL) (400-700 nm) lies outside of the spectral range The mechanism by which VL induces skin damage appears to be,
of what photobiologists historically define as deleterious radiation. at least in part, attributable to inducing oxidative damage to skin
Visible light comprises approximately 40% of the solar radiation cells. Using in vitro methods, it was determined that VL exposure
that reaches the surface of the earth. Only a few studies have leads to micronuclei formation without cyclobutane pyrimidine
evaluated the effects of the visible light range of wavelengths dimers (a hallmark of UVB damage), which is associated with the
on skin. Considering that during outdoor activities, skin is exposed induction of oxidative DNA damage in skin cells.31 The peak wave-
to the full solar spectrum, including visible light; this research is length range for this DNA base modification has been determined to
quite relevant. Visible light can cause various dermatologic condi- be between 400 and 450 nm.32 In vitro and ex vivo studies have also
tions such as solar urticaria, chronic actinic dermatitis, phototoxic established the pro-inflammatory, ROS-generating effects of visible
and photoallergic skin reactions, porphyrias, erythema, and pigmen- light. Irradiation of human skin equivalents with visible light induced
tation induction. Detailed studies carried out by Kollias et al production of ROS, pro-inflammatory cytokines, and matrix metallo-
‫برنزه‬ showed that VL can induce skin pigmentation, which was sus- proteinase (MMP)-1 expression. Using clinical models to assess the
27
tained for up to 10 weeks. It has also been demonstrated, using generation of free radicals from oxidative stress, higher levels of free
a xenon-mercury lamp, that VL (up to 470 nm) can induce imme- radical activity were found after visible light exposure.33 Exposure to
28
diate pigment darkening (IPD) similar to that of UVA1. The peak VL also generated hydrogen peroxide in a dose-dependent fashion.
for the IPD response was shown to be between wavelengths 300 Pretreatment of cells with antioxidants prevented this free radical
and 500 nm,29 closer to the UVA1 range as might be expected. generation in skin cells by VL, suggesting a potential modality of pre-
Visible light has been shown to induce erythema and a tanning vention of VL damage to skin. In addition to the MMP-1 induction,
response in dark skin, but not in fair-skinned individuals.23 Skin VL has also been shown to increase pro-inflammatory cytokines (IL-
type IV-VI responded to VL by increased skin pigmentation while 1 alpha, IL-1 receptor antagonist, IL-6, GM-CSF, and IL8), induction
skin type II showed no visible response to VL.25 In addition to of MMP-1 and -9, and activation of EGFR/ERK pathway.33
pigmentation, VL also induced skin erythema. This response was In terms of current knowledge on protection against VL, com-
greater for darker skin types than lighter skin types. This is likely mercially available sunscreens were found to have minimal effects
due to the higher amount of chromophores (eg, melanin) in darker on reducing visible light-induced ROS, suggesting that traditional
skin than lighter skin. The melanin absorbs light-generating heat UVA/UVB sunscreens do not protect the skin from visible light-
25
causing vasodilation, followed by skin erythemal response. induced responses. There is a need to identify strategies that can be
MCDANIEL ET AL. | 129

successfully employed in the future to achieve more comprehensive stimulation of melanin production.41 Recent work demonstrates that
34
photoprotection of human skin beyond ultraviolet radiation. IR and heat exposure each induce cutaneous angiogenesis and
This need has been further heightened by recent research exam- inflammatory cellular infiltration, disrupt the dermal extracellular
ining high-energy visible light (HEVL), or blue light, which ranges in matrix by inducing matrix metalloproteinases, and alter dermal struc-
wavelength from 400 to 500 nm. Previously, blue light had been pri- tural proteins, thereby contributing to premature skin aging.38 IR-A
marily known as a therapeutic treatment option for several cuta- radiation, in addition to UV, has been demonstrated to alter the col-
neous disorders.35 More recently, data showing blue light causing lagen equilibrium of the dermal extracellular matrix in at least 2
ROS-mediated photoaging, inflammation, and genetic expression ways: (a) by leading to an increased expression of the collagen-
changes in markers, such as MMP1 and IL8, have been published. degrading enzyme matrix metalloproteinase 1 and (b) by decreasing
Additional data demonstrate the ability of antioxidants such as toco- the de novo synthesis of the collagen itself. IR-A-induced expression
pherol to block or prevent this ROS-mediated blue light skin dam- of matrix metalloproteinase-1 (MMP-1) was found to be mediated
age.33,36 Most recently, blue light has been studied as it relates to by the formation of intracellular reactive oxygen species (ROS). It
pigmentation and the inducible generation of the tyrosinase-dopa- was demonstrated in vivo that exposure of normal skin to IR-A radi-
chrome tautomerase complex in type III-VI melanocytes, which ation caused upregulation of MMP-1 mRNA expression by a factor
causes sustained tyrosinase activity. The key sensor for melanocyte of 3 to 4 times.34 In response to IR irradiation, mitogen-activated
detection of blue light is thought to be Opsin-3 (OPN3), a calcium- protein kinase signaling pathways were activated mediating the
dependent protein that can activate tyrosinase activity, and other upregulation of matrix metalloproteinase-1 expression.7
37
pathways/mechanisms such as CAMKII, CREB, p38, and MITF. Paradoxically, low-level IR-A has been shown to have stimulatory
HEVL has been overlooked as a photoaging causative agent as evi- effects on cellular mechanisms and even collagen production,
denced by the fact that of the 40 000 skincare products launched in through a nonthermal process known as photobiomodulation. This
35
2016, only 9 claimed to have HEVL/blue light protection. effect has been demonstrated in cultured human fibroblasts by vary-
ing ratios of wavelengths in a dual wavelength LED system com-
prised of 595/870 nm wavelengths and a total energy fluence of
4 | PART III: HEAT, INFRARED LIGHT, AND 0.1 J/cm2.42 Additionally, low-level exposure to IR-A has been
SKIN AGING shown to have increased healing benefits as well as application in
preconditioning the skin to be better prepared for upcoming insult
Infrared (IR) radiation consists of wavelengths from 800 nm to or trauma. Known as photoprevention, this process has been shown
1 mm, and it is subdivided into 3 regions of increasing wavelength, in laboratory experiments, but mimics the natural world as the red
IR-A (700-1400 nm), IR-B (1400-3000 nm), and IR-C (3000 nm- and IR-A wavelengths present in early morning sunlight may have
1 mm). Given that almost half of the solar energy reaching the evolved to ready the skin for later, more deleterious UVR as the day
earth’s surface is in the IR range, solar IR is expected to have signifi- progresses.43 Uncontrolled, frequent, or long-term exposure to near-
38
cant biological effects on human skin. In direct sunlight, the tem- infrared can also cause thermal burns and aging effects such as
perature of human skin rises to about 40◦C following the “Bakers Arms” or “Glassblowers Face.”38
conversion of absorbed IR into heat. Skin temperature rise is also While there are some similarities in the cutaneous biological
higher in skin of color as compared to fair skin.23 So far, our knowl- effects of IR-A and UV radiation exposure, there are also significant
edge of the effects of IR radiation or heat on skin aging is limited. differences. The cellular response to IR-A irradiation involves the
IR-B and IR-C do not penetrate deeply into the skin, although mitochondrial electron transport chain.40,44 IR-A-irradiated cells
they cause increases in skin temperature. More than 65% of the revealed an increase in mitochondrial superoxide anion production.
more highly energetic IR-A reaches the dermis and subcutaneous The treatment of fibroblasts with the mitochondrial targeted antioxi-
layers without increasing skin temperature.38 Human skin is increas- dant MitoQ completely abrogated the IR-A, but not the UVB or
ingly exposed to IR-A radiation; most relevant sources are (i) natural UVA1 response.45 ROS relevant for IR-A-induced signaling originated
solar radiation consisting of over 30% IR-A, (ii) artificial IR-A sources from the mitochondrial electron transport chain, because (i) chemical
used for therapeutic or wellness purposes, and (iii) artificial UV inhibition of the electron transport chain prevented IR-A, but not
sources contaminated with IR-A. As part of natural sunlight, IR-A sig- UVB or UVA1, radiation-induced MMP-1 expression, and (ii) peroxi-
nificantly contributes to extrinsic skin aging.39 Recent studies have some proliferator-activated receptor gamma coactivator-1 (PGC-1),
demonstrated that the solar radiation which induces skin damage overexpressing fibroblasts with increased electron transport chain
can occur not only from ultraviolet (UV) radiation alone, but also content were hypersensitive to IR-A radiation-induced gene expres-
from longer wavelengths, in particular near-infrared radiation (IR-A sion. When healthy human subjects were exposed to physiologically
radiation, 700-1400 nm).40 relevant doses of IR-A, eighty percent of the tested individuals
Different skin types react differently to IR-A. Lightly pigmented responded to IR-A radiation by upregulation of MMP-1 expression.
skin is most vulnerable with reduction in collagen synthesis and Specifically, IR-A irradiation caused increased expression of MMP-1
increase in MMP-1 and MMP-3 expression, while highly pigmented in the dermis, but not in the epidermis. Raman spectroscopy
skin has minimal effect on collagen and MMPs, but demonstrates revealed that IR-A radiation also caused a significant decrease in the
130 | MCDANIEL ET AL.

antioxidant content of human skin. In vitro studies had previously stress via the formation of lipid peroxidation products. Chronic expo-
shown that IR-A-induced MMP-1 expression was mediated through sure to high levels of atmospheric ozone has been shown to induce
an oxidative stress response, which originates from the mitochon- antioxidant depletion, as well as oxidation of lipids and proteins,
drial electron transport chain. In addition, IR-A also causes the stimu- within the outermost skin layer (stratum corneum) and the lung res-
lation of extracellular signal-regulated kinase 1/2 activation in human piratory tract lining fluids (RTLFs).51 O3 can react readily with protein
7
dermal fibroblasts. IR-A induced a variety of genes at the transcrip- nucleophiles via Michael addition and Schiff base formation. The tar-
tome level in primary human skin fibroblasts. Microarray analysis gets of O3 are cysteine, histidine, and lysine residues of protein, and
revealed 599 IR-A-regulated transcripts. The IR-A-induced transcrip- affect lipids of skin by 4-hydroxynonenal-protein adducts (HNE
tome differed from changes known to be induced by UV. IR-A- adduct) formation and skin lipid peroxidation1 (Figure 4). In addition
responsive genes included the following categories: extracellular to damaging cellular lipids and proteins, O3 can also deplete skin of
matrix, calcium homeostasis, stress signaling, and apoptosis.46 Using its essential antioxidants. Studies in hairless mice SKH-1 demon-
RT-PCR technology and chemical inhibitors for the various signaling strated that when exposed to 0.8 ppm of O3, the depletion of alpha-
pathways, ERK1/2, p38, JNK, PI3K/AKT, STAT3, and IL-6, as well as tocopherol in lung and plasma, and the induction of heme oxygenase
the calcium-mediated signaling pathways, were identified as specifi- 1, cyclooxygenase 2, and proliferation of cell nuclear antigen in both
cally targeted by IR-A. This IR-A gene response is triggered by mito- skin and lung were observed. O3-exposed animals showed a similar
chondrial and, to a lesser extent, nonmitochondrial production of extent of upregulation of COX-2 and PCNA in lung and skin,
reactive oxygen species.46 In addition to MMP-1, IR and heat also whereas HO-1 was more responsive in skin than in lung (sevenfold
stimulate the expression of tropoelastin and fibrillin-1, 2 main com- induction vs. twofold induction). This study demonstrated
ponents of elastic fibers, and matrix metalloproteinase (MMP)-12, that skin exposure to high levels of O3 not only affects antioxidant
the most active MMP against elastin, in human skin in vivo. This levels and oxidation markers in the SC, but also induces stress
suggests that the abnormal production of tropoelastin and fibrillin by responses in the active layers of the skin, most likely by indirect
heat in human skin, and their degradation by various MMPs, such as mechanisms, as it is unlikely that O3 itself penetrates the protective
MMP-12, may contribute to the accumulation of elastotic material in SC layers.52 Murine skin also responded to O3 by an increase in 4-
photoaged skin.40 hydroxynonenal-protein adducts and a rapid upregulation of HSP27
Protection from heat and IR-A has been demonstrated by pre- (20-fold), and more delayed induction of HSP70 (2.8-fold) and heme
treatment of skin with antioxidants, N-acetyl cysteine or genistein, oxygenase-1 (fivefold). O3 exposure also led to the induction of
for 24 h prior to heat or IR-A.47 It has also been demonstrated nitric oxide synthase (iNOS) 6-12 h following the exposure. This
in vitro that the effects of IR-A can be abolished by incubation of study suggested that skin exposure to high levels of O3 not only
cultured human dermal fibroblasts or treatment of human skin with affects antioxidant levels and oxidation markers in the SC, but also
specific antioxidants.48 Thus, heat and IR-A irradiation most likely induces stress responses in the active layers of the skin.53
promote premature skin aging and skin elastosis, and topical applica- In vitro studies using cultured keratinocytes have demonstrated
tion of appropriate antioxidants may represent an effective photo- the damaging effects of O3 on skin cells and the protective effects of
protective strategy. antioxidants against the O3-induced skin damage.54 O3 exposure
induced cytotoxicity, inhibited cellular proliferation, and increased the
formation of 4-hydroxynonenal (HNE) protein adducts. Furthermore,
5 | PART IV: OZONE AND SKIN AGING O3 decreased activation of the redox-sensitive transcription factor NF-
kB, which is involved in transcribing pro-inflammatory cytokines, and
Ozone (O3) is considered one of the most toxic environmental stres- therefore constitutes one of the main players associated with O3-
sors to which humans are continuously exposed. The human skin, par- induced skin inflammation. Cells exposed to O3 demonstrated a dose-
ticularly the upper layer of the epidermis, acts as an effective barrier dependent increase in p65 subunit nuclear expression as a marker of
between the body and the environment, but can also be a major tar- NF-kB activation. NF-kB is a master regulator of a variety of cellular
get of air pollutants, making the skin one of the most susceptible tis- responses responsible for apoptosis signaling pathways, cell prolifera-
sues to the oxidative damaging effect of O3. The average tion, angiogenesis, metastasis, tumor promotion, and inflammation.
tropospheric amount of O3 is generally less than 0.08 ppm, much Therefore, O3 can indirectly activate pathways in skin that can be dam-
lower than in the stratosphere (10 ppm). Yet, in large metropolises aging to the skin as well as the whole organism. Indeed, direct activa-
such as Mexico City, as well as European cities such as Rome, Milan, tion of O3 on the skin cell NF-kB has been demonstrated.49 O3 also
and Paris, O3 can reach very high toxic concentrations, especially dur- activates the pro-inflammatory cytokine IL-8 expression in skin.54
49
ing the summer. Anthropogenic emissions, mainly of nitrogen oxi- Additional downstream effects of O3 in skin include activation of ker-
des, as well as methane, carbon monoxide, and sulfuric compounds, atin 10, PCNA gene, stress proteins such as heat shock proteins, HO1,
have caused a progressive increase in O3 concentration up to and AhR (aryl hydrocarbon receptor), and pro-inflammatory markers
0.8 ppm or more in the last 30 years.50 (COX 2 and iNOS).55 Pretreatment of keratinocytes with a topical vita-
While O3 is not able to penetrate the skin, the damage induced min C compound mixtures prevented the aforementioned O3-induced
by O3 is mainly a consequence of its ability to induce oxidative cytotoxicity in skin keratinocytes and in 3D skin models.54
MCDANIEL ET AL. | 131

6 | PART V: SMOG AND OTHER


PARTICULATE MATTER AND SKIN AGING

Epidemiological studies suggest a correlation between increased air-


borne particulate matter (PM) and adverse health effects. A study
on the combined effects of 3 ambient air pollutants, ozone (O₃),
nitrogen dioxide (NO₂), and fine particulate matter (PM) with a
median aerodynamic diameter of less than 2.5 lm (PM (2.5)), corre-
lated the short-term changes in AQHI (Air Quality Health Index)
with emergency department (ED) visits for urticaria in Windsor-area
hospitals in Canada. Urticaria is among the skin pathologies com-
monly associated with pollution. Positive and significant correlations
were observed between AQHI levels and OR or ED visits for urti-
caria in Windsor. This study clearly demonstrated associations
between ambient air pollution and urticarial, confirming that air pol-
lution affects skin conditions.57 A recent epidemiological study
showed a link between traffic-related air pollution and the forma-
tion of lentigines in Caucasian and Asians populations.58 Soot,
which frequently results from diesel exhaust, is a mixture of carbon
particles covered by organic compounds including polycyclic aro-
FIGURE 4 Ozone effects on human skin matic hydrocarbons. Polycyclic aromatic hydrocarbon-induced acti-
vation of aryl hydrocarbon receptor signaling in epidermal cells
might provide a mechanistic explanation for these epidemiological
In an epidemiological study conducted in Shanghai, China, a observations.59 Indeed, stimulation of primary human epidermal ker-
time series analysis was conducted to evaluate the association atinocytes with ambient soot was recently shown to cause an acti-
between emergency room (ER) visits for skin conditions and O3 vation of the aryl hydrocarbon receptor and subsequent gene
exposure. A variety of skin conditions including urticaria, eczema, transcription.60
contact dermatitis, rash⁄other nonspecific eruption, infected skin The mechanisms of PM health effects are believed to involve
disease, and other skin diseases were correlated with daily air pol- oxidative stress and inflammation. To gain further insight, the as to
lution data obtained from the Shanghai Monitoring Centre, includ- the effects of PM on skin, investigations were conducted using
ing O3, particulate matter with aerodynamic diameter of 10 lm or reconstituted human epidermis (RHE). RHE was exposed to concen-
less (PM10), sulfur dioxide (SO2), and nitrogen dioxide (NO2). trated ambient particles (CAPs) (25 or 100 lg/ml) for 24 or 48 hours.
There were direct correlations only between environmental O3 A local reactive oxygen species (ROS) production increase, presum-
content and the skin conditions.56 These findings are biologically ably generated from metals present on the particle, contributed to
plausible due to the propensity of O3 to be highly reactive, with observed lipid oxidation. Furthermore, as a consequence of altered
a high oxidizing power, which can react with biomolecules in the redox status, NF-kB nucleus translocation was increased upon CAPs
skin to form ozonides and free radicals. This study provides evi- exposure, as well as an increase in cyclooxygenase 2 and cyto-
dence that the current atmospheric level of O3 has an adverse chrome P450 levels, which may be involved in the inflammatory
effect on the skin health of the general population and strength- response initiated by PM. CAPs also triggered an apoptotic process
ens the rationale to further investigate the harmful effect of O3 in skin.61 Transition electron microscopy revealed that CAPs were
on human skin, as well as to evaluate possible measures to coun- able to penetrate skin tissues. These findings contribute to the
teract its effect. understanding of the cutaneous pathophysiological mechanisms initi-
A recent in vivo clinical study further validated the O3-induced ated by CAPs exposure, where oxidative stress and inflammation
skin damage that was observed in vitro and in 3D skin models. O3 may play predominant roles.
exposure increased epidermal levels of 4-hydroxynonenal (4HNE)
and 8-iso Prostaglandin F2 alpha (8-iso PGF2a), confirming the O3
interaction with skin lipids. In addition, after O3 exposure, there was 7 | PART VI: THERAPEUTIC INTERVENTION
a clear and significant increase in NF-kB p65 subunit activation that OF ATMOSPHERIC SKIN AGING
paralleled the increased levels of cyclooxygenase-2 (COX-2) and
metalloproteinase-9 (MMP-9). Of note, after O3 exposure, there was Photoprotection of human skin by means of sunscreens or daily
a dramatic decrease in both types I and III collagen (Col I and III). skincare products is traditionally centered on the prevention of acute
Pretreatment with topical vitamin C compound mixtures was able to (eg, sunburn) and chronic (eg, skin cancer and photoaging) skin dam-
prevent the O3-induced skin damage. (details in Section 6). age that may result from exposure to ultraviolet rays (UVB and
132 | MCDANIEL ET AL.

UVA). Within the last decade, however, it has been appreciated that as a secondary antioxidant. Lipoic acid is both water- and lipid-solu-
wavelengths beyond the ultraviolet spectrum, in particular visible ble and acts as an essential cofactor for several enzyme systems. It
light and infrared radiation, contribute to skin damage. As a result, is able to regenerate (reduce) primary antioxidants such as vitamin C,
an effort has been made to develop skincare products that not only glutathione, and vitamin E. N-acetyl cysteine, a thiol amino acid
protect against UVA or UVB radiation, but provide photoprotection derivative, which can also regenerate GSH, is another example of a
against visible light and infrared radiation as well. The role of exoge- secondary antioxidant.3
nously supplied antioxidants or natural compounds possessing high Antioxidant enzyme systems are those enzymes that can regen-
antioxidant capacity has been evaluated. erate antioxidants. These include the following: GSH peroxidases,
GSH reductase, glutathione S-transferases (GSTs), and other
enzymes that directly neutralize ROS, such as superoxide dismutases
7.1 | Skin endogenous antioxidant systems:
(SODs) catalases and quinone reductases. Many of these enzymes
Human skin is equipped with an array of antioxidants and enzyme are reduced in aging and photoaged skin. Some of these enzymes
systems to protect the cells from damaging effects of free radicals require metal cofactors such as Cu, Mn, and Zn for SOD and Se for
(Table 2). Antioxidant molecules such as vitamin A, vitamin C, and catalase. Providing bioavailable metals to skin can thus boost their
vitamin E slow the process of aging either by preventing free radicals activities and provide improved antioxidant protection to skin.
from oxidizing sensitive biological molecules or by reducing the for- Although some metal ions are cofactors of antioxidant enzymes,
mation of free radicals and quenching the already formed ROS.18 In metal ions can also act as pro-oxidants. For example, under certain
addition, enzymes such as superoxide dismutase, (SOD), catalase, conditions, Fe and Cu can cause oxidation reactions in skin and dam-
and glutathione biosynthesizing enzymes protect the tissues from age proteins and cell organelles (eg, Fenton reaction catalyzed by
free radicals. The levels of these antioxidants, as well as antioxidant Fe2+ ion). Therefore, metal chelating agents that can neutralize tran-
enzymes, are reduced by intrinsic and extrinsic factors such as age sition metals and prevent their production of free radicals in skin can
and atmospheric aggressors. Replenishing these antioxidants either also be considered secondary antioxidants. In addition, several small
by topical application or by dietary supplementation can protect skin metabolites generated within the skin (uric acid, urocanic acids, urea,
from ROS.5,6 several natural amino acid derivatives etc.) can also act as potent
Primary or free radical scavenging antioxidants inhibit oxidation antioxidants. The endogenous (biological) antioxidants present in the
via chain terminating reactions. Inhibition occurs by transfer of a skin (vitamin C, E, GSH, lipoic acid) show a gradient in human epider-
proton to the free radical species. Examples of primary antioxidant mis (high levels in basal layers to low levels in upper layers). The
molecules include glutathione (GSH), alpha-tocopherol, (vitamin E levels of these antioxidants decrease with age and photooxidation,
and derivatives), and ascorbic acid (vitamin C). GSH and ascorbic acid thus providing a rationale for the supplementation of these antioxi-
are water-soluble, whereas vitamin E and biquinols are fat-soluble dants topically in cosmetic formulations. Glutathione, however, is a
and membrane-bound. Because the ability of different antioxidants tripeptide, and its ionic charges would make it an unlikely candidate
to interact with each other creating the already described “antioxi- for substantial percutaneous absorption.62 Therefore, further
dant network,” the combined use of more than one molecule has research is required to substantiate the effectiveness of GSH.
been suggested.51 Secondary antioxidants are often used in combi- Solar ultraviolet (UV) radiation is one of the most significant
nation with primary antioxidants to yield synergistic stabilization environmental DNA-damaging agents to which humans are exposed
effects. They protect primary antioxidants from degradation. In constantly. Sunlight, specifically UVB and UVA, triggers various types
human skin, lipoic acid (an organic sulfur containing molecule) serves of DNA damage. DNA damage may have several deleterious conse-
quences, such as cell death, mutagenesis, photoaging, and cancer.
UVA and UVB photons can not only be directly absorbed by DNA
T A B L E 2 Endogenous antioxidants present in human skin creating direct damage, but also by other chromophores leading to
Antioxidant compounds Antioxidant enzyme systems the formation of reactive oxygen species, which may indirectly
Glutathione (water-soluble) Glutathione peroxidases cause DNA damage.63 The current preventative measures, such as
Vitamin C (water-soluble) Glutathione transferases sunscreens and topical antioxidants, have not shown to be com-

Tocopherols (Vitamin E) (lipid-soluble) Glutathione reductase pletely effective in blocking the effects of UV radiation.
Topical application of DNA repair enzymes serves to supplement
Ubiquinone (lipid-soluble) Superoxide dismutases
(Cu/Zn and Mn) intrinsic DNA repair mechanisms. Several DNA repair enzymes criti-
Lipoic acid (water- and lipid-soluble) Catalase cal to the prevention of cutaneous nonmelanoma cancers have been

Carotenoids (lycopene, lutein, Heme oxygenase isolated and added to topical preparations designed for skin cancer
zeaxanthin, beta-carotene)—all prevention. These DNA repair enzymes can enhance DNA re-
from dietary sources pair and provide a more efficient response to carcinogenesis and sun
Uric acid, urocanic acid, and other Quinone reductase damage.64 UV-induced DNA damage in the epidermis (and to some
minor components of amino acid extent in the dermis) occurs mainly as UV-induced cross-linked DNA
metabolites (N-acetyl cysteine) in skin
base pairs and the formation of cyclobutane pyrimidine dimers
MCDANIEL ET AL. | 133

(CPD’s), 6-4 photoproducts (6-4PPs), and 8-oxo-20 deoxyguanosine. properties. Polyphenols comprise several families of compounds
The DNA repair enzymes consist of photoylases (enzymes that including tannins (gallic acid esters of glucose or other sugars), lig-
remove CPDs), endonucleases (breakdown the intra-DNA base pairs nins, and flavonoids. Flavonoids include several thousand com-
or base excision enzymes), and 8-oxoguanine DNA glycosylase- pounds, among them the flavonols, flavones, catechins, flavanones,
OGG1 (repair of 80 HdG).65 Topical preparations containing these anthocyanidins, and isoflavonoids.69,70 A list of commonly used
DNA repair enzymes have been successfully used for patients with polyphenols derived from natural sources is shown in Table 3. In
nonmelanoma skin cancer and Xeroderma pigmentosum66 and actinic addition to pure polyphenolic compounds, a variety of phytoextracts,
67
keratosis. Topical creams containing these enzymes encapsulated rich in polyphenols, have also been used in cosmetic products for
in liposomes have been used as a successful strategy for treatment protection from sun damage. A major challenge in topical antioxidant
and reversal of these skin conditions. These treatment options supplementation lies in providing the optimal formulation.71 An
resulted in decreased solar elastosis, reduction in actinic keratosis effective formula must be optimized for compatibility (between
and improvement in lentigines, smoothness, moisture and overall antioxidants and with other formulation components), stability, pene-
appearance.68 tration, and dosage. In general, antioxidants such as vitamin C are
unstable in neutral and alkaline pH, and are stable at or below the
pKa of the acid form (pH 3.5 or lower). Vitamin C can also be stabi-
7.2 | Exogenously supplied topical antioxidants for
lized by other antioxidants such as ferulic acid; this strategy is used
skin protection
to keep high vitamin C formulas stable. Using appropriate surfac-
Plant-derived compounds such as carotenoids and polyphenols are tants, emollients, and solvents to enhance the penetration of antioxi-
an important group of compounds showing excellent antioxidant dants in the skin is a common strategy to enhance the skin
availability of antioxidants. The optimum concentration of an antioxi-
T A B L E 3 Polyphenols and their natural sources commonly used dant is another critical variant. The “higher the better” approach
in Skincare products to protect skin from photodamage does not always apply with topical formulations. Stability, bioavail-

Polyphenol compounds Natural sources ability, and the inclusion of supplemental ingredients all impact the
optimal concentration of an antioxidant that can be incorporated in
Resveratrol Red grape skin, peanuts, mulberry,
blueberry, and bilberry. Red wine a formulation. There is evidence that the skin absorption of vitamin
contains significant amounts C is only dose-dependent up to a certain point, and increasing the
Epigallocatechin-3-gallate Green tea, black tea concentration beyond that has been shown to affect the formula-
(EGCG) and other epicatechins tion’s stability. The inclusion of antioxidants in formulations, specifi-
Caffeine, caffeic acid, quinic acid, Grains, fruits coffee fruit cally sunscreens, has been a topic of interest in recent years. It is
chlorogenic acid, and ferulic
well known that pH-dependent antioxidants, such as vitamin C, can-
acid
not be effectively combined with sunscreen formulations. Recent
Quercetin Fruits, leafy vegetables, berries,
studies indicate that select combinations of antioxidants in sunscreen
tomato broccoli etc.
formulas have evidence of clinical efficacy. Further developments in
Phloretin and its glucoside Apple skin
phloridzin this area are still required to understand the parameters for antioxi-
dant usage.
Genistein, daidzein, and their Soya extract
derivatives
Rosmarinic, ursolic and carnosic Sage, rosemary, lavender, oregano 7.3 | Clinical studies using topical antioxidant
acids and other pentacyclic thyme, peppermint, and other
triterpenes herbs
protection against atmospheric skin damage
Silymarin Milk thistle UV-induced erythema can be prevented in human skin by the topical
Curcuminoids From the root of Curcuma longa application of vitamin C compound mixtures. The pretreatment of an
(turmeric) antioxidant mixture consisting of 10% L-ascorbic acid + 2%
Isothiocyanates (sulforaphane) Broccoli sprouts and other similar Phloretin + 0.5% Ferulic Acid has been shown to inhibit erythema
vegetables
induced by 1-5 MED of solar UV radiation.70 UV exposure resulted
Ellagic acid Pomegranate in erythema, sunburn cell formation, thymine dimer formation,
Chlorogenic acid Green coffee beans and fruit MMP-9 expression, and p53 protein expression induction. These sun
Glycyrrhizic acid Licorice damaging effects were suppressed by the topical pretreatment of a
Polyphenols comprise of thousands of natural, plant-derived compounds. vitamin C compound mixture 5 days prior to UV exposure.
They are generally classified as hydrolysable tannins (gallic acid esters of A similar approach using vitamin C compound mixtures (MIX1
sugars) and phenylpropanoids such as lignins, flavonoids, and condensed (15% L-ascorbic acid + 1% Alpha-tocopherol + 0.5% Ferulic Acid)
tannins. Flavonoids include thousands of compounds including flavonols,
and MIX2 (10% L-ascorbic acid + 2% Phloretin + 0.5% Ferulic Acid)
flavones, catechins, flavanones, anthocyanidins, and isoflavonoids. The
following table is a partial list of these antioxidant compounds that are was successful in reducing the ozone-induced damage in human skin
used in cosmetic compositions Oresajo et al.6 in vivo.72 The forearms of subjects were exposed to O3 (0.8 ppm)
134 | MCDANIEL ET AL.

for 5 days (3 hr per day for 5 consecutive days), which caused the scientific information which are guiding future therapies. The gaps in
induction of pro-inflammatory cytokines, NF-kB expression, p65 sub- current protective measures are clear, and innovative new
unit activation, and increased levels of COX2 and MMP-9 with a approaches are required to address these issues. The concept of
concomitant decrease in type I and III collagen in skin. On the sites inside/outside protection from environmental exposure requires fur-
of the forearm pretreated with the vitamin C compound mixtures, ther exploration, as well as how to produce sustained protection
the damage caused by O3 was inhibited. These results suggest that throughout daily exposure. As direct protective measures are not
topical treatment with vitamin C compound mixtures can help pre- available or are impractical for some of the emerging light and pollu-
vent the noxious effects of ozone in skin. tion damage, the role of antioxidants is a vital part of defending
Vitamin C compound mixtures (MIX1 (15% L-ascorbic acid + 1% against this damage. In addition to topical antioxidant treatment, oral
Alpha-tocopherol + 0.5% Ferulic Acid) and MIX2 (10% L-ascorbic antioxidant therapies have also been shown to be effective in pre-
acid + 2% Phloretin + 0.5% Ferulic Acid) were also proven effective venting sun damage to skin. The benefits of a multicomponent nutri-
in preventing IR-A-induced skin damage. Pretreatment of human skin tional supplement on photodamaged skin were recently
in vivo with the vitamin C compound mixtures significantly demonstrated.76 Twenty-four-week dietary supplementation with a
decreased IR-A-induced MMP-1 expression.34 Photoaging may be multicomponent nutritional supplement improved the skin quality
prevented and treated with a variety of modalities, including combi- index in healthy 35- to 65-year-old women with Fitzpatrick skin
73
nation therapies comprised of topical antioxidants and sunscreens. types I-IV. A number of studies have also demonstrated the effec-
A combination of SPF 30 sunscreen with and without an antioxidant tiveness of antioxidants on photoprotective benefits when given
cocktail containing grape seed extract, vitamin E, ubiquinone, and orally.77–80
vitamin C was evaluated for protection of human skin against IR-A New in vivo testing methods to evaluate the content as well as
radiation-induced MMP-1 upregulation.74 As expected, exposure to the efficacy of antioxidants on human skin are being developed.81
IR-A radiation significantly upregulated MMP-1 expression, as com- Direct detection of reactive oxygen species (ROS) has recently been
pared to unirradiated skin, and this response was significantly developed for evaluating antioxidant efficacy in vivo, under physio-
reduced with the application of the SPF30 sunscreen plus the logical conditions over time.82 Successful evaluation of ROS species
antioxidant cocktail prior to IR-A radiation. In contrast, treatment of and scavenging of these molecules via fluorescence measurements
human skin with the SPF30 sunscreen alone did not provide signifi- of tape strips to measure skin’s natural antioxidant defense capabil-
cant protection. ity,83 and the efficacy of topically applied antioxidants, have been
The common denominator that links skin damage to the different developed.84 New fluorescent probes are being developed to reveal
solar wavelengths is the enhanced production of reactive molecule the presence of the ROS on tape strips of stratum corneum.
species (RMS) within the skin cells followed by increased oxidative In the area of ingredient development, novel phytoextracts with
stress. A clinical study which evaluated a combination of broad-spec- improved antioxidant activity are continuously explored. New syn-
trum sunscreen and select antioxidants, that provide protection from thetic antioxidants with sunscreen booster capabilities are being
total radiation while promoting skin repair, was conducted.75 In vitro developed to provide UV protection from outside, as well as antioxi-
studies have indicated this combination could play a role in prevent- dant protection from the inside of the skin.41 Plant stem cells pre-
ing the formation of UV-induced sunburn cells and cyclobutane sent a novel approach and show promise for future formulations.
pyrimidine dimers while preserving or improving the expression of Antioxidant conjugates with anti-aging peptides are patented for skin
ECM genes. In addition, ex vivo studies suggest the combination applications.85,86 New formulations (eg, liposomes) and novel deliv-
may prevent IR-A-triggered fragmentation of elastin fibers and ery options (eg, iontophoresis or carrier molecules) for targeted and
expression of MMP-1. Initial clinical studies indicate that this broad- specific delivery are being evaluated.
spectrum sunscreen in combination with select antioxidants reduces In addition, combinations of multiple antioxidants and chemical
the increase in surface temperature seen with IR radiation. A signifi- components have been designed with the intent of providing protec-
cant improvement in the appearance of lines and wrinkles was tion for all types of ROS to provide complete protection. This com-
reported as early as week 2 in patients using this blend. This study plex was tested both in vitro and in vivo in a series of tests. First,
suggests that select antioxidants along with SPF protection can help the measurement of ROS in skin explants following application of
counterbalance the deleterious effects of free radicals on skin cells the new complex, another current AOX serum, and saline controls. A
by promoting endogenous repair.75 standard peroxide assay was completed in 2 separate trials on the
samples, and it was determined that there was a reduction of 53%
and 41% peroxide in the new complex relative to the current AOX
8 | PART VII: FUTURE TRENDS serum (P = .01, test 1; P = .005, test 2, respectively). The complex
was also tested for minimal erythemal dose (MED) in 5 human sub-
New approaches for therapeutic intervention of atmospheric skin jects. Two 5 cm 9 5 cm sites were marked on the lower back, one
damage are emerging. While our current understanding of the full was randomized to be treated by the AOX complex for 4 days and
impact atmospheric aggressors have on premature skin aging is still the other served as untreated control. The treated and untreated
incomplete, recent discoveries are providing compelling new sites were irradiated with 19, 29, and 39 each subject’s MED
MCDANIEL ET AL. | 135

(determined for each individually previously). Digital images, redness 13. Bernstein EE, Brown DB, Schwartz MD, Kaidbay K, Ksenzenko SM.
measurements, and 3 mm punch biopsies were collected from each The polyhydroxy acid gluconolactone protects skin against UV in an
in vitro model of cutaneous photodamage. Dermatol Surg.
subject for analysis of biomarkers. The AOX complex-treated sites
2006;30:189-196.
demonstrated 94%, 76%, and 50% less UV-induced erythema at 19, 14. Trautinger F. Mechanisms of photodamage to skin and its func-
29, and 39 MED (P = .025, P < .001, and P = .004, respec- tional consequences for skin aging. Clin Exp Dermatol. 2001;26:
tively).Additionally, there was skin illustrated by reductions in thy- 573-577.
15. Kulms D, Shwartz T. Molecular mechanisms of UV induced apopto-
mine dimers (P < .02), MMP-9 (P < .005), p53, and sunburn cells.
sis. Photoderm Photoimm Phtomed. 2000;16:195-201.
Education of both the public and the medical community about 16. Krutmann J, Schroeder P. Role of mitochondria in photoaging of
these dangers will be required. Overall, the future focus of skincare, human skin – the defective powerhouse model. J Invest Dermatol.
with regard to combatting the effects of atmospheric skin aging, will be 2009;14:44-49.
17. Gilchrest BA, Park HY, Eller MS, Yaar M. Mechanisms of ultraviolet
the development of novel evaluation technologies and the use of
light-induced pigmentation. Photochem Photobiol. 1996;63:1-10.
potent and stable topical antioxidants both to prevent and repair dam- 18. Briganti S, Picardo M. Antioxidant activity, lipid peroxidation and
age. skin diseases: what is new. J Eur Acad Dermatol Venerol 2003;17:
663-669.
19. Fisher GJ, Kang S, Varani J, et al. Mechanisms of photoaging and
ACKNOWLEDGMENT chronological skin aging. Arch Dermatol. 2002;138:1462-1470.
20. Senftleben U, Karin M. The IKK/NF-kappaB pathway. Crit Care Med.
The authors would like to thank Sewon Kang MD, MPH, FAAD for 2002;30(suppl 1):S18026.
his contributions to the visible light segment of this manuscript, and 21. Schwarz T, Luger TA. Effect of UV irradiation on epidermal cell cyto-
Kumar Pillai PhD for his critical review and editorial assistance with kine production. Photochem Photobiol 1989;4:1-13.
22. Fisher GJ, Voorhees JJ. Molecular mechanisms of photoaging and its
this manuscript.
prevention by retinoic acid: ultraviolet irradiation induced MAP
kinase signal transduction cascades that induce AP-1 regulated
matrix metalloproteinases that degrade human skin in vivo. J Investig
ORCID Dermatol Symp Proc. 1998;3:61-68.
23. Sklar LR, Almutawa F, Lim HW, Hamzavi I. Effects of ultraviolet radi-
David McDaniel http://orcid.org/0000-0002-7595-5456
ation, visible light, and infrared radiation on erythema and pigmenta-
tion: a review. Photochem Photobiol Sci. 2013;12:54-64.
24. Grimes PE. Management of hyperpigmentation in darker racial ethnic
REFERENCES groups. Semin Cutan Med Surg. 2009;28:77-85.
25. Mahmoud BH, Ruvolo E, Hexsel CL, et al. Impact of long wavelength
1. Valacchi G, Sticozzi C, Pecorelli A, Cervellati F, Cervellati C, Maioli E. UVA and visible light on melanocompetent skin. J Invest Dermatol.
Cutaneous responses to environmental stressors. Ann N Y Acad Sci. 2010;130:2092-2097.
2012;1271:75-81. 26. Marionnet C, Nouveau S, Hourblin V, et al. UVA-1 induced skin
2. Krutmann J, Bouloc A, Sore G, Bernard BA, Passeron T. The skin darkening is associated with molecular changes even in highly pig-
aging exposome. J Dermatol Sci. 2017;85:152-161. mented skin individuals. J Investig Dermatol 2017;137:1184.
3. Pinnell SR. Cutaneous photodamage, oxidative stress, and topical 27. Kollias N, Bager A. An experimental study of the changes in pigmen-
antioxidant protection. J Am Acad Dermatol. 2003;48:1-19. tation in human skin in vivo with visible and near infrared light. Pho-
4. Bikers DR, Athar M. Oxidative stress in the pathogenesis of skin dis- tochem Photobiol. 1984;39:651-659.
ease. J Invest Dermatol. 2006;126:2565-2575. 28. Rosen CF, Jacques SL, Stuart ME, Gange RW, et al. Immediate
5. Thiele JJ, Schroeter C, Hsieh SN, Podda M, Packer L. The antioxi- pigment darkening: visual and reflectance spectrophotometric
dant network of the stratum corneum. Curr Probl Dermatol. 2001; analysis of action spectrum. Photochem Photobiol. 1990;51:583-
29:26-42. 588.
6. Oresajo C, Pillai S, Yatskayer M, Puccetti G, McDaniel D. 29. Pathak RF, Riley FC, Fitzpatrick TB. Melanogenesis in human skin
Antioxidants and Skin aging: a review. Cosmetic Dermatol. 2009;22: following exposure to long-wave ultraviolet and visible light. J Invest
563-568. Dermatol. 1962;39:435-443.
7. Schieke SM, Schroeder P, Krutmann J. Cutaneous effects of infrared radi- 30. Randhawa M, Seo I, Liebel F, Southall MD, Kollias N, Ruvolo E. Visi-
ation: from clinical observations to molecular response mechanisms. Photo- ble light induces melanogenesis in human skin through a photoadap-
dermatol Photoimmunol Photomed. 2003;19:228-234. tive response. PLoS ONE. 2015;10:e0130949.
8. Matsumu Y, Ananthaswamy HN. Toxic effects of ultraviolet radiation 31. Hoffmann-Dorr S, Greinert R, Volkmer B, Epe B. Visible light
on the skin. Toxicol Appl Pharmacol. 2004;195:298-308. (>395 nm) causes micronuclei formation in mammalian cells without
9. Tewari A, Grage MM, Harrison GI, Sarkany R, Young AR. UVA1 is generation of cyclobutane pyrimidine dimers. Mutat Res
skin deep: molecular and clinical implications. Photochem Photobiol 2005;572:142-149.
Sci. 2013;12:95-103. 32. Kielbassa C, Roza L, Epe B. Wavelength dependence of oxidative
10. Wang F, Smith NR, Tran BA, Kang S, Voorhees JJ, Fisher GJ. Dermal DNA damage induced by UV and visible light. Carcinogenesis
damage promoted by repeated low-level UV-A1 exposure despite 1997;18:811-816.
tanning response in human skin. JAMA Dermatol. 2014;150:401-406. 33. Liebel F, Kaur S, Ruvolo E, Kollias N, Southall MD. Irradiation
11. Damian DL, Matthews YJ, Phan TA, Halliday GM. An action spec- of skin with visible light induces reactive oxygen species and matrix-
trum for ultraviolet radiation-induced immunosuppression in humans. degrading enzymes. J Invest Dermatol. 2012;132:1901-1907.
Br J Dermatol. 2011;164:657-659. 34. Grether-Beck S, Marini A, Jaenicke T, Krutmann J. Photoprotection
12. Kang S, Fisher GJ, Voorhees JJ. Photoaging: pathogenesis, preven- of human skin beyond ultraviolet radiation. Photodermatol Photoim-
tion and treatment. Clin Geriat Med. 2001;17:643-659. munol Photomed. 2014;30:167-174.
136 | MCDANIEL ET AL.

35. Mendrok-Edinger C, Campiche R, Gadsinki K, Schuetz R. Into the 57. Kousha T, Valacchi G. The air quality health index and emergency
blue: novel test reveals blue light damage, protection strategies. Cos- department visits for urticaria in Windsor, Canada. J Toxicol Environ
metics Toiletries. 2018;133:12-29. Health A. 2015;78:524-533.
36. Nakashima Y, Ohta S, Wolf AM. Blue light induced oxidative stress 58. Hu € tter A, Gao W, et al. Traffic-related air pollution con-
€ls A, Vierko
in live skin. Free Radic Biol Med. 2017;108:300-310. tributes to development of facial lentigines: further epidemiological
37. Regazetti C, Sormani L, Debayle D, et al. Melanocytes sense blue evidence from Caucasians and Asians. J Investig Dermatol.
light and regulate pigment through Opsin-3. J Invest Dermatol. 2016;136:1053-1056.
2018;138:171-178. 59. Krutmann J, Liu W, Li L, et al. Pollution and skin: from epidemiologi-
38. Cho S, Shin MH, Kim YK, et al. Effects of infrared radiation and heat cal and mechanistic studies to clinical implications. J Dermatol Sci.
on human skin aging in vivo. J Investg Dmermatol 2009;14:15-19. 2014;76:163-168.
39. Schroeder P, Haendeler J, Krutmann J. The role of near in- 60. Nakamura M, Morita A, Seite0 S, Haarmann-Stemmann T, Grether-
frared radiation in photoaging of the skin. Exp Gerontol. Beck S, Krutmann J. Environment-induced Lentigines (EILs): forma-
2008;43:629-632. tion of solar lentigines beyond ultraviolet radiation. Exp Dermatol
40. Schroeder P, Calles C, Benesova T, Macaluso F, Krutmann J. Photo- 2015;24:407e11.
protection beyond ultraviolet radiation–effective sun protection has 61. Magnani ND, Muresan XM, Belmonte G, et al. Skin damage mecha-
to include protection against infrared A radiation-induced skin dam- nisms related to airborne particulate matter exposure. Toxicol Sci.
age. Skin Pharmacol Physiol. 2010;23:15-17. 2016;149:227-236.
41. Velasco MV, Sarruf FD, Salgado-Santos IM, Haroutiounian-Filho CA, 62. Lin JY, Selim MA, Shea CR, et al. UV photoprotection by combina-
Kaneko TM, Baby AR. Broad spectrum bioactive sunscreens. Int J tion topical antioxidants vitamin C and vitamin E. J Am Acad Derma-
Pharm. 2008 Nov 3;363:50-57. tol. 2003;48:866-874.
42. McDaniel DH, Weiss RA, Geronemus RG, Mazur C, Wilson S, Weiss 63. Schuch AP, Moreno NC, Schuch NJ, Menck CF. Garcia CC Sunlight
MA. Varying ratios of wavelengths in dual wavelength LED photo- damage to cellular DNA: focus on oxidatively generated lesions. Free
modulation alters gene expression profiles in human skin fibroblasts. Radic Biol Med. 2017;107:110-124.
Lasers Surg Med 2010;42:540-545. 64. Kabir Y, Seidel R, Mcknight B, Moy R. DNA repair enzymes: an
43. Barolet D, Christiaens F, Hamblin MR. Infrared and skin: friend or important role in skin cancer prevention and reversal of photodam-
foe. J Photochem Photobiol, B. 2016;155:78-85. age–a review of the literature. J Drugs Dermatol. 2015;14:297-303.
44. Schroeder P, Calles C, Krutmann J. Prevention of infrared-A radia- 65. Emanuele E, Spencer JM, Braun M. From DNA repair to proteome
tion mediated detrimental effects in human skin. Skin Therapy Lett. protection: new molecular insights for preventing non-melanoma
2009;14:4-5. skin cancers and skin aging. J Drugs Dermatol. 2014;13:274-281.
45. Schroeder P, Pohl C, Calles C, Marks C, Wild S, Krutmann J. Cellular 66. Yarosh D, Klein J, O’Connor A, Hawk K, Rafal E, Wolf P; Xeroderma
response to infrared radiation involves retrograde mitochondrial sig- Pigmentosum Study Group. Effects of topically applied T4 endonu-
naling. Free Radic Biol Med. 2007;43:128-135. clease 5 in liposomes on skin cancer in xeroderma pigmentosum. A
46. Calles C, Schneider M, Macaluso F, Benesova T, Krutmann J, Schroe- randomized study. Xeroderma pigmentosum study group. Lancet.
der P. Infrared a radiation influences the skin fibroblast transcrip- 2001;7:926.
tome: mechanisms and consequences. J Investig Dermatol. 67. DeBoyes T, Kouba D, Ozog D, et al. Reduced number of actinic ker-
2010;130:1524-1536. atosis with topical application of DNA repair enzyme Creams. JDD.
47. Chen Z, Seo JY, Kim YK, et al. Heat modulation of tropoelastin, fib- 2010;9:1519.
rillin-1, and matrix metalloproteinase-12 in human skin in vivo. J 68. Spencer JM, Morgan MB, Trapp KM, Moon SD. Topical formulation
Investig Dermatol 2005;124:70-78. engendered alteration in p53 and cyclobutane pyrimidine dimer
48. Schroeder P, Lademann J, Darvin ME, et al. Infrared radiation- expression in chronic photodamaged patients. JDD. 2012;12:336-
induced matrix metalloproteinase in human skin: implications for 340.
protection. J Invest Dermatol. 2008;128:2491-2497. 69. Svobodova A, Psotova J, Walterova D. Natural phenolics in the pre-
49. Doche C, Dufour G, Foret G, et al. Summertime tropospheric-ozone vention of UV induced skin damage – A review. Biomed Papers.
variability over the Mediterranean basin observed with IASI. Atmos 2003;14:137-158.
Chem Phys. 2014;14:10589-10600. 70. Oresajo C, Stephens T, Hino PD, et al. Protective effects of a topical
50. Mustafa MG. Biochemical basis of ozone toxicity. Free Radical Biol antioxidant mixture containing vitamin C, ferulic acid and phloretin
Med. 1990;9:245-265. against UV induced photodamage in human skin. J Cosmetic Derma-
51. Packer L, Valacchi G. Antioxidants and the response of skin to oxida- tol 2008;7:290-297.
tive stress: vitamin E as a key indicator. Skin Pharmacol Appl Skin 71. Oresajo C, Pillai S, Manco M, Yatskayer M, McDaniel D. Antioxi-
Physiol 2002;15:282-290. dants and the skin: understanding formulation and efficacy. Derma-
52. Valacchi G, Pagnin E, Corbacho AM, et al. In vivo ozone exposure tology Therapy. 2012;25:252-259.
induces antioxidant/stress-related responses in murine lung and skin. 72. Valacchi G, Pecorelli A, Belmonte G, et al. Protective effects of topi-
Free Radic Biol Med. 2004;36:673-681. cal vitamin C compounds mixtures against ozone induced damage in
53. Valacchi G, van der Vliet A, Schock BC, et al. Ozone exposure acti- human skin. J Investig Dermatol. 2017;137:1373.
vates oxidative stress responses in murine skin. Toxicology. 73. Han A, Chien AL, Kang S. Photoaging. Dermatol Clin. 2014;32:291-
2002;179:163-170. 299.
54. Valacchi G, Sticozzi C, Belmonte G, et al. Oresajo C vitamin C com- 74. Grether-Beck S, Marini A, Jaenicke T, Krutmann J. Effective photo-
pound mixtures prevent ozone-induced oxidative damage in human protection of human skin against infrared A radiation by topically
keratinocytes as initial assessment of pollution protection. PLoS applied antioxidants: results from a vehicle controlled, double-blind,
ONE. 2015;10:e0131097. randomized study. Photochem Photobiol. 2015;91:248-250.
55. Valacchi G, Fortino V, Bocci V. The dual action of ozone on the skin. 75. McDaniel DH, Hamzavi IH, Zeichner JA, et al. Total defense + repair:
Br J Dermatol. 2005;153:1096-1100. a novel concept in solar protection and skin rejuvenation. J Drugs
56. Qiao L, Cai J, Wang H, et al. PM2.5 constituents and hospital emer- Dermatol. 2015;14:s3-s11.
gency-room visits in Shanghai, China. Environ Sci Technol. 76. Birnbaum JE, McDaniel DH, Hickman J, Dispensa L, Le Moigne A,
2014;48:10406-10414. Buchner L. A multicenter, placebo-controlled, double-blind clinical
MCDANIEL ET AL. | 137

trial assessing the effects of a multicomponent nutritional supple- 83. Ziosi P, Besco E, Vertuani S, Solaroli N, Manfredini S. A non-invasive
ment for treating photoaged skin in healthy women. J Cosmet Der- method for the in vivo determination of the skin antioxidant capac-
matol. 2017;16:120-131. ity (IAC-S). Skin Res Technol. 2006;12:303-308.
77. Fern andez-Garcıa E. Skin protection against UV light by dietary an- 84. Cornacchione S, Sadick NS, Neveu M, et al. In vivo skin antioxidant
tioxidants. Food Funct. 2014;5:1994-2003. effect of a new combination on a specific vitis vinifera shoot extract
78. Baliga MS, Katiyar SK. Chemoprevention of photocarcinogenesis by and a biotechnological extract. J Drugs Dermatol. 2007;6:s8-s13.
selected dietary botanicals. Photochem Photobiol Sci. 2006;5:243-253. 85. Majeed M, Renukeshwar CH, Nagabhushanam K. Peptides modified
79. Katiyar SK. Interleukin-12 and photocarcinogenesis. Toxicol Appl with triterpenoids and small organic molecules: synthesis and use in
Pharmacol. 2007;224:220-227. cosmeceuticals, US patent # 8383594, 2013.
80. Aust O, Stahl W, Sies H, Tronnier H, Heinrich U. Supplementation 86. Majeed M, Nagabhooshanam K. Oleanoyl peptide composition and
with tomato-based products increases lycopene, phytofluene, and method of treating skin aging, US patent # 8987212, 2015.
phytoene levels in human serum and protects against UV-light-
induced erythema. Int J Vitam Nutr Res. 2005;75:54-60.
81. McDaniel DH, Neudecker BA, Dinardo JC, Lewis JA 2nd, Maibach
HI. Idebenone: a new antioxidant – Part I. Relative assessment of How to cite this article: McDaniel D, Farris P, Valacchi G.
oxidative stress protection capacity compared to commonly known Atmospheric skin aging—Contributors and inhibitors. J
antioxidant. J Cosmet Dermatol. 2005;4:10-17.
Cosmet Dermatol. 2018;17:124–137.
82. Khabiri F, Hagens R, Smuda C, et al. Noninvasive monitoring of
oxidative skin stress by ultraweak photon emission (UPE)-measure- https://doi.org/10.1111/jocd.12518
ment. I: mechanisms of UPE of biological materials. Skin Res Technol.
2008;14:103-111.
Copyright of Journal of Cosmetic Dermatology is the property of Wiley-Blackwell and its
content may not be copied or emailed to multiple sites or posted to a listserv without the
copyright holder's express written permission. However, users may print, download, or email
articles for individual use.

You might also like