ArticlePDF AvailableLiterature Review

Abstract

The effects of environmental insults on human health are a major global concern. Some of the most noxious pollutants that humans are exposed to include ozone (O3), particulate matter (PM), and cigarette smoke (CS). Since the skin is the first line of defense against environmental insults, it is considered one of the main target organs for the harmful insults of air pollution. Thus, there is solid evidence that skin pathologies such as premature aging, atopic dermatitis (AD), and psoriasis are associated with pollutant exposure; all of these skin conditions are also associated with an altered redox status. Therefore, although the mechanisms of action and concentrations of O3, PM, and CS that we are exposed to differ, exposure to all of these pollutants is associated with the development of similar skin conditions due to the fact that all of these pollutants alter redox homeostasis, increasing reactive oxygen species production and oxidative stress. A main product of oxidative stress, induced by exposure to the aforementioned pollutants, is 4‐hydroxy‐2‐nonenal (HNE), which derives from the oxidation of ω‐6 polyunsaturated fatty acids. HNE is a highly reactive compound that can form adducts with cellular proteins and even DNA; it is also an efficient cell signaling molecule able to regulate mitogen‐activated protein kinase pathways and the activity of redox‐sensitive transcription factors such as Nrf2, AP1, and NFκB. Therefore, increased levels of HNE in the skin, in response to pollutants, likely accelerates skin aging and exacerbates existing skin inflammatory conditions; thus, targeting HNE formation could be an innovative cosmeceutical approach for topical applications.
REVIEW ARTICLE
Involvement of 4-hydroxy-2-nonenal in pollution-induced skin
damage
Alessandra Pecorelli
1
| Brittany Woodby
1
| Roxane Prieux
2
| Giuseppe Valacchi
1,2,3
1
Plants for Human Health Institute,
Department of Animal Sciences, North
Carolina State University, Kannapolis,
North Carolina
2
Department of Life Sciences and
Biotechnology, University of Ferrara,
Ferrara, Italy
3
Department of Food and Nutrition, Kyung
Hee University, Seoul 02447, Korea
Correspondence
Giuseppe Valacchi, Department of Animal
Sciences, Plants for Human Health Institute,
NC Research Center, North Carolina State
University, Kannapolis, NC.
Email: gvalacc@ncsu.edu, giuseppe.
valacchi@unife.it
Funding information
Marie Sklodowska-Curie, Grant/Award
Number: 765602; European Unions
Horizon 2020
Abstract
The effects of environmental insults on human health are a major global concern.
Some of the most noxious pollutants that humans are exposed to include ozone (O
3
),
particulate matter (PM), and cigarette smoke (CS). Since the skin is the first line of
defense against environmental insults, it is considered one of the main target organs
for the harmful insults of air pollution. Thus, there is solid evidence that skin patholo-
gies such as premature aging, atopic dermatitis (AD), and psoriasis are associated with
pollutant exposure; all of these skin conditions are also associated with an altered
redox status. Therefore, although the mechanisms of action and concentrations of O
3
,
PM, and CS that we are exposed to differ, exposure to all of these pollutants is associ-
ated with the development of similar skin conditions due to the fact that all of these
pollutants alter redox homeostasis, increasing reactive oxygen species production and
oxidative stress. A main product of oxidative stress, induced by exposure to the afore-
mentioned pollutants, is 4-hydroxy-2-nonenal (HNE), which derives from the oxida-
tion of ω-6 polyunsaturated fatty acids. HNE is a highly reactive compound that can
form adducts with cellular proteins and even DNA; it is also an efficient cell signaling
molecule able to regulate mitogen-activated protein kinase pathways and the activity
of redox-sensitive transcription factors such as Nrf2, AP1, and NFκB. Therefore,
increased levels of HNE in the skin, in response to pollutants, likely accelerates skin
aging and exacerbates existing skin inflammatory conditions; thus, targeting HNE for-
mation could be an innovative cosmeceutical approach for topical applications.
KEYWORDS
ozone, particulate matter, cigarette smoke, peroxidation
1|INCREASING AIR POLLUTION
AND IMPLICATIONS FOR HUMAN
SKIN HEALTH
Ubiquitous exposure to environmental insults is a major
global concern for public health. Over the past 30 years, the
damaging effects of pollutants on cutaneous tissue have
become an area of growing research interest, also in view of
increased environmental pollution. Indeed, a recent search in
PubMed using the keywords skin and pollutionreveals
Abbreviations: AD, atopic dermatitis; AhR, aryl hydrocarbon receptor;
AP1, activator protein-1; HNE, 4-hydroxy-2-nonenal; CO, carbon
monoxide; COX2, cyclooxygenase-2; CS, cigarette smoke; DEP, diethyl
phthalate; DHN, 1,4-dihydroxy-2-nonene; DnBP, di(n-butyl)phthalate;
MAPKs, mitogen-activated protein kinases; NFκB, nuclear factor κB;
NO
2
, nitrogen dioxide; Nrf2, nuclear factor (erythroid-derived 2)-like 2; O
3
,
ozone; PA, protein adducts; PM, particulate matter; PPARs, peroxisome-
proliferator-activated receptors; PUFA, polyunsaturated fatty acids; RHEs,
reconstituted human epidermis tissues; SC, stratum corneum; SO
2
, sulfur
dioxide; SRB1, scavenger receptor class B type I; UFPs, ultrafine
particulates; VOCs, volatile organic compounds.
Alessandra Pecorelli and Brittany Woodby contributed equally to this study.
Received: 21 February 2019 Revised: 19 March 2019 Accepted: 2 April 2019
DOI: 10.1002/biof.1513
© 2019 International Union of Biochemistry and Molecular Biology
BioFactors. 2019;112. wileyonlinelibrary.com/journal/biof 1
more than 2000 papers related to this topic (https://www.ncbi.
nlm.nih.gov/pubmed/?term=skin+pollution). Until the year
2000, only a few articles were published on this issue; how-
ever, in the last 20 years, interest in this field has grown expo-
nentially, reaching approximately 100 manuscripts per year.
This increased interest is related not only to the proven and
well-established worsening of air quality but also to the evi-
dence of a direct correlation between skin pathologies and air
pollution exposure.
14
Indeed, the skin is the first line of
defense because it is the interface between the environment
and our body. Therefore, cutaneous tissue is considered, along
with eyes, lung, brain, and digestive tract, as one of the main
target organs for the harmful insults of air pollution.
5
1.1 |Cutaneous tissues as a gateway for the
outdoor pollutants
Growing evidence demonstrates that the skin also constitutes
a route of entry for ambient pollutants in the human body,
promoting considerable systemic consequences in almost all
internal organs.
68
In a very elegant series of experiments,
Weschler et al. (2015) were able to demonstrate the ability
of volatile organic compounds (VOCs), specifically diethyl
phthalate (DEP) and di(n-butyl)phthalate (DnBP), to be
absorbed by the skin. Surprisingly, the amounts of phthalate
metabolites found in urine were similar when the subjects
were exposed to pollutants either by breathing or by dermal
exposure, suggesting that the ability of our body to absorb
pollutants from the outdoor environment by the cutaneous
tissues is similar, if not more efficient than the respiratory
tract. Even more striking is the finding that dermal absorp-
tion of VOCs increases tremendously with age; indeed, sub-
jects over 60 years old had five times higher levels of the
phthalates in urine compared to 30-year-old subjects.
8
Interestingly, a role of clothing in influencing the cutaneous
uptake of harmful compounds from polluted air has been inves-
tigated by several groups
911
; surprisingly, clean clothes are
only able to protect the skin by about 2030% from pollution
absorption, eventually being a source of pollutant accumulation,
and increasing cutaneous uptake if not changed daily.
9,10
2|POLLUTION AND SKIN
PATHOLOGIES
The use of the word pollutioncan be misleading given that
there are several different pollutants that can affect our
health. In addition, not all of them have the same concentra-
tion in the air and the same mechanism of action. Based on
their chemical and physical properties as well as their
sources, the United States Environmental Protection Agency
has identified the most common air pollutants, also known as
criteria air pollutants, as ozone (O
3
), particulate matter
(PM), carbon monoxide (CO), lead, sulfur dioxide (SO
2
),
and nitrogen dioxide (NO
2
) (https://www.epa.gov/criteria-
air-pollutants). Clear evidence of the correlation between
each single pollutant and skin disorders has not yet been
established; however, the noxious effects of O
3
, cigarette
smoke (CS), and PM have been well demonstrated, as
described in the following section.
Overall, there is solid evidence that pathologies such as
atopic dermatitis (AD), psoriasis, acne, and, in some cases,
also skin cancer can be associated with pollutant exposure. In
particular, exposure to O
3,
PM, NO
2,
and CS has been demon-
strated to be associated with cutaneous pathologies. The semi-
nal epidemiological work by Xu et al. (2011) demonstrated
the association of these pollutants with cutaneous pathologies
by analyzing the association between emergency-room (ER)
visits for skin conditions and levels of air pollutants including
O
3
,PM
10
,SO
2
, and NO
2
. During 2 years of sampling, over
68,000 visits to the ER for skin disorders were recorded, and a
clear correlation between O
3
concentration and cutaneous
issues was demonstrated. In particular, the authors underlined
how several skin conditions such as urticaria, eczema, contact
dermatitis, rash/other nonspecific eruption, and infected skin
diseases were exacerbated when the subjects were exposed to
increased levels of this pollutant.
12
These data suggest a role
of O
3
in inducing inflammatory skin pathologies. Another
more recent publication has further examined the association
of short-term changes in air quality with emergency depart-
ment (ED) visits for urticaria in Canada. A total of 2905 ED
visits were analyzed, and a positive and significant correlation
was observed between air quality levels and ED visits for
urticaria, confirming that air pollution can affect skin
physiology.
13
AD is a chronic and recurrent cutaneous inflammatory dis-
ease that begins in the early stage of life. The pathogenesis of
ADisusuallylinkedtoskinbarrier alteration and immune dys-
regulation.
14,15
Indeed, changes in the stratum corneum (SC)
composition, which is the outermost layer of the skin, can facili-
tate the penetration of allergens that can then be associated with
the development of AD.
15
As opposed to the outside-in model of
AD pathogenesis,
16
the inside-out theory suggests that Th2 cyto-
kines are able to modulate the expression of proteins present in
the SC, thereby disrupting the skin barrier.
17
Although there is still existing controversy between these
two theories, one fact is clear; the perturbation of the skin
barrier plays a key role in AD, and this perturbation can be
induced by environmental pollutant exposure. For instance,
there is now evidence that air pollution influences the preva-
lence of AD. In a fairly recent study, it was shown that, in a
population of almost 5000 children from France, there was a
direct correlation between the development of eczema and
pollution levels (PM
10
and NOx).
18
In addition, a study con-
ducted in the Munich metropolitan area revealed a strong
2PECORELLI ET AL.
positive association between the distance to the nearest main
road and eczema; in particular, it was found that NO
2
was
positively associated with eczema in children exposed to
traffic-related air pollution.
19
In a more recent work, PM
10
and NO
2
exposure during the first trimester of pregnancy
was associated with the development of infantile AD.
20
It
has also been demonstrated that maternal smoking during
pregnancy and/or in the first year after birth is a major risk
factor for the development of AD among children aged
between 6 and 13 years.
21
Similarly, fetal tobacco smoke
exposure during the third trimester of pregnancy was posi-
tively associated with a higher cumulative incidence of
atopic eczema/dermatitis syndrome in exposed infants in a
Japanese study; the authors suggest that maternal smoking
might induce epigenetic changes in the fetal allergen-specific
immune responses, promoting development of AD.
22
Thus, the evidence that exposure to environmental tobacco
smoke during early childhood can predispose children to later
development of AD has been documented, but it is still not
clear whether current smokers develop AD. This point was
well clarified in the paper by Lee et al., in which, among
83 patients diagnosed with adult-onset AD, more than 50%
were current smokers, and about one-third have smoked in
the past.
23
This study strongly supports the idea of the associ-
ation between current smoking and the development of adult-
onset AD, as well as a correlation between exposure to CS
and AD in nonsmokers.
Besides eczema and AD, psoriasis is another inflammatory-
related skin disease that appears to be associated with air pollu-
tion. Indeed, it has been proposed that exposure to pollutants
such as PM,
24
CS,
25
or O
326
can activate the aryl hydrocarbon
receptor (AhR), and this can further activate Th17 cells
27
;the
main cells involved in psoriasis and present in psoriatic lesions.
Although there is controversy in the link between CS and psori-
asis, a recent study with over 17 million patients over the age of
20 years that were followed for 8 years was able to clearly show
the positive correlation between risk of psoriasis and smoking
period.
28
In addition, this correlation was stronger for subjects
that smoked more than two packs per day and much lower for
0.5 pack smokers.
28
Interestingly, CS has been also connected with the devel-
opment of acne,
4
another multifactorial skin disease. Acne is
usually characterized by increased sebum production, abnor-
mal keratinization of the pilosebaceous duct, and inflamma-
tion driven by the presence of Propionibacterium acnes. It
has been shown that these processes are induced by an
altered redox status, which pollutant exposure can gener-
ate.
29
Indeed, the ability of PM, CS, and even O
3
to increase
reactive oxygen species (ROS) production and activate a
cascade of events, leading to increased OxInflammation, has
been well proven. In a recent work, our laboratory was able
to show that CS-induced OxInflammation hampers the
ability of sebocytes to uptake cholesterol via oxidation of an
important skin receptor, the scavenger receptor class B type
I (SRB1).
30
This pathway has also been observed in other
skin models, such as keratinocytes and 3D skin equivalents,
and is related not only to CS but also to PM and O
3
exposure
3133
(GV unpublished data).
In addition to the aforementioned skin conditions, environ-
mental changes, due to rapid industrialization and urbanization
of the last few decades, are suspected to be the main drivers of
the increased incidence of skin pigmentation in geographic
regions with very heavy pollution, such as India and South
East Asia. Indeed, pollutants, such as PM and polycyclic aro-
matic hydrocarbons (PAH), due to their ability to enter the skin
via nanoparticles, also appear to be important risk factors for
facial hyperpigmentation disorders, specifically melasma.
34
Exposure to PM has also been implicated in the development,
persistence, and exacerbation of other cutaneous conditions,
such as AD, acne, psoriasis, skin aging, androgenetic alopecia,
and skin cancer.
35
In conclusion, ambient air pollutants, such as PM, CS, or
O
3
, seem to be involved in the pathogenesis of inflammatory
skin diseases (e.g., AD, acne, and psoriasis) via a common
denominator, that is, through enhancing oxidative stress and
proinflammatory mediators (OxInflammation phenomena)
36
(Figure 1).
3|MECHANISMS INVOLVED IN
POLLUTION AFFECT CUTANEOUS
TISSUES
3.1 |Atmospheric skin damage
Although all pollutants are able to induce OxInflammation as
the final outcome of their harmful effects, it is interesting to
note that each of them can affect human skin physiology
through different mechanisms of action. Based on their chem-
ical and physical characteristics, only some air contaminants
are able to penetrate through the layers of the skin, reaching
the dermis. Therefore, the potential damaging effects and the
way by which each pollutant impacts skin structure and func-
tion differs substantially.
O
3
is a small molecule and strong oxidizing agent that
directly acts on the surface of cutaneous tissue, disseminating
its detrimental effects into the deeper epidermal layers through
the generation of a cascade of ozonation products. Although it
is not a radical species per se, O
3
is able to oxidize compo-
nents of the cell membrane, mainly lipids, generating classical
radical species such as hydroxyl radicals that, in turn, drive
the production of cytotoxic, nonradical species including alde-
hydes. Due to its high reactivity and chemical and physical
properties of low aqueous solubility within the skin, O
3
is not
able to reach and directly damage live epidermal and dermal
PECORELLI ET AL.3
cells. In fact, it is well proven that this pollutant is entirely
consumed through reaction with skin surface lipids and the
intercellular lipids of the SC.
37
As a mechanically protective and flexible structure mainly
constituted of anucleated deadcorneocytes, the outermost
stratum of our skin is enriched by sebaceous and intercellular
lipids including squalene, triglycerides, ceramides, free fatty
acids, wax monoesters, and cholesterol. Since all of these
lipids are easily prone to oxidation, as a natural consequence,
their reaction with O
3
can generate several secondary messen-
gers able to trigger signaling cascades across the different
layers of skin, leading to prooxidative and proinflammatory
processes.
3840
For instance, in a recent study, it has been shown that O
3
exposure increased levels of HNE in human skin, and this
correlated with increased proinflammatory markers such as
COX2 and NFκB.
41
Interestingly, the levels of HNE followed
a clear gradient pattern, with high levels in the upper epider-
mis and lower levels in the dermis, suggesting that the effect
of O
3
on the skin is indeed mediated by oxidation products
generated mainly in the upper layers of the epidermis.
41
Therefore, it is possible to claim that the effect of O
3
on
cutaneous tissues is a consequence of its reaction with the
lipids present in the SC.
This concept was first advanced by Pryor et al.
42
in rela-
tion to the respiratory tract, suggesting that exposure of non-
cellular constituents of surface epithelial cells to O
3
is capable
of generating potentially toxic peroxidation products. Extrap-
olation of this concept to cutaneous tissues suggests that O
3
reacts directly with the SC lipids that contribute to the cutane-
ous tissue protective barrier,
39
generating products that are
able to penetrate the SC and target keratinocytes. It is con-
cluded that O
3
not only affects cutaneous antioxidantlevels
and oxidation markers in the SC but also induces cellular
responses in the deeper layers of the skin.
Low-molecular-weight antioxidants are present in high
concentrations, especially in the epidermis. Oxidative stress
can overwhelm skin defenses and increase the formation of
oxidized cell components. Topical exposure to tropospheric
O
3
induces oxidative imbalances in the skin. Oxidative dam-
age to the SC may result in barrier perturbation and in the
production of lipid oxidation products that can act as sec-
ond messengersin the deeper layers of the skin, which, in
turn, elicits repair responses and/or the induction of defense
proteins such as NRF2 and/or heat shock proteins (HSPs).
Oxidative injury to the outermost layers of the skin can
initiate localized inflammatory responses, resulting in the
recruitment of phagocytes and their tightly regulated, cell-
specific NAD(P)H-oxidase systems for generating oxidants,
further amplifying the oxidative stress damage.
41
As of today, the potential overall mechanism by which O
3
is
able to affect skin has been described. It is generally understood
that the toxic effects of O
3,
although it is not a radical species
per se, are mediated through free radical reaction either directly
by the oxidation of biomolecules to give classical radical spe-
cies (hydroxyl radical) or by driving the radical-dependent
production of cytotoxic, nonradical species (aldehydes). Fur-
thermore, the formation of oxidation products, characteristic of
damage from free radicals, has been shown to be prevented by
the addition of the vitamins E and C. O
3
is not able to penetrate
the SC, so it first interacts with the lipids present in the outer-
most layer of the skin, leading to the generation of a number of
bioreactive species. Our lab together with other recent works
has provided some evidence that these bioactive compounds are
likely to penetrate the underlying cutaneous tissues, as demon-
strated by the presence of several proinflammatory markers in
the deeper layer of the skin.
40
It can be suggested that reaction
with the well-organized interstitial lipids and protein constitu-
ents of the outermost SC barrier, and diffusion of bioreactive
products from this tissue into the viable layers of the epidermis,
may represent a contribution to the development/exacerbation
of skin disorders associated with O
3
exposure
.
Indeed, once
these mediatorsare able to reach live cells (keratinocytes,
fibroblasts, etc.), they can induce a cellular defensive and
FIGURE 1 Consequences of 4-hydroxynonenal (HNE)
production in response to pollutants. Exposure of the skin to ozone,
cigarette smoke, and particulate matter induces lipid peroxidation and
production of HNE. This product of lipid peroxidation can form
covalent bonds with the histidine, cysteine, and lysine residues of
proteins, such as cytochrome c, through Michael addition, generating
ROS and oxidative stress, which can promote/exacerbate cutaneous
conditions such as psoriasis, premature aging, and AD. ROS, reactive
oxygen species
4PECORELLI ET AL.
inflammatory response that leads to an inflammatory/oxidative
vicious cycle, OxInflammation. This, unless quenched by
endogenous or exogenous mechanisms, will damage the skin
and compromise its barrier functions, contributing to extrinsic
skin aging.
3.2 |Mechanisms involved in CS effects
on skin
CS is a highly complex aerosol composed of more than 4700
chemicals and consists of a gas phase and a particulate phase.
Mainstream smoke (the combination of inhaled and exhaled
smoke after taking a puff of a lit cigarette) includes particu-
lates suspended in a gaseous phase. It is widely recognized
that CS contains high levels of prooxidants,
43
with more than
10
14
low molecular weight carbon- and oxygen-centered rad-
icals per puff present in gas-phase smoke.
44
Sidestream
smoke goes into the air directly from a burning cigarette and
is the main component of second-hand smoke. The chemical
constituents of sidestream smoke are different from those of
directly inhaled (mainstream) CS; it has been shown that
inhaled sidestream CS is approximately four times more
toxic per gram of total particulate matter than mainstream
CS.
45
Furthermore, sidestream condensate, compared to
mainstream, is about three times more toxic per gram and
two to six times more tumorigenic per gram. The gas/vapor
phase of sidestream smoke is responsible for most of the sen-
sory irritation and respiratory tract epithelium damage.
45
As mentioned above, the toxic effect of CS on the skin
has been well demonstrated. Exposure to CS can result in
impaired wound healing, development of squamous cell car-
cinoma, oral cancer, acne, psoriasis, eczema, hair loss, and
premature skin aging.
46
Epidemiological studies strongly
correlated CS to premature skin aging.
4749
Moreover, the
obvious esthetic damage of the skin by CS was well docu-
mented by Dr. Model more than 30 years ago, who defined
the so-called smoker's face,characterized by grayskin
(smoker's melanosis) and deep wrinkles (smoker's wrin-
kle).
50
Indeed, wrinkle formation is a typical feature associ-
ated with tobacco smoking.
51
CS is able to affect skin aging
by activating MMPs in the connective tissues.
52
For instance,
MMP-1 induces the degradation of both collagen and elastic
fibers. In addition, production of the procollagen types I and
III is affected by CS, while MMP-1 and MMP-3 are strongly
induced.
53
The mechanisms involved in CS-induced skin
aging remain unresolved, although it is believed that activa-
tion of the AhR signaling pathway contributes to this effect.
CS contains water-insoluble PAHs, which have been linked
to activation of the AhR signaling pathway. AhR is involved in
the regulation of development, hypoxia signaling, and circadian
rhythms, and belongs to a family of proteins that reside in the
cytoplasm in an inactive complex with accessory proteins.
54,55
Once activated, AhR dissociates from some of the proteins in
the inactive complex and translocates to the nucleus, where it
dimerizes with Arnt.
56
The AhR/Arnt heterodimer activates the
transcription of xenobiotic-metabolizing genes
57,58
;someof
which encode proteins involved in growth control, cytokines,
nuclear transcription, and regulators of extracellular matrix pro-
teolysis.
59,60
Therefore, the AhR pathway may be involved in
the effects of tobacco smoke on skin. In support of this idea, CS
increased MMP-1 mRNA induction in primary keratinocytes
and fibroblasts, and AhR knockdown abolished this effect,
suggesting the involvement of AhR activation in extrinsic skin
aging induced by CS.
61
In addition to premature aging, CS has also been linked to
psoriasis. As previously mentioned, a recent study of over
17 million subjects demonstrated a positive correlation between
smoking and psoriasis, which correlated with how many packs
per day the subjects smoked.
28
The molecular basis of this
effect is likely due to increased oxidative stress in the skin
induced by CS. In fact, our lab has demonstrated that CS expo-
sure in keratinocytes increases NAPDH oxidase activity as
assessed via p47 and p67 membrane translocation, resulting in
increased H
2
O
2
levels and mitochondrial superoxide produc-
tion.
31
We also observed that CS exposure in keratinocytes
increases the levels of HNE and acrolein adducts.
31
We believe
that increased NAPDH oxidase activity is due to increased pro-
duction of HNE adducts in response to CS exposure, since
Yun et al. (2005) demonstrated that HNE production is able to
directly activate NOx.
62
Moreover, the increased levels of
superoxide anion or H
2
O
2
produced by NOx can regulate the
AhR signaling pathway, connecting AhR activation to oxida-
tive stress responses. It is also possible that the AhR transcrip-
tion factor itself can be modified by HNE. Since increased
oxidative stress in the skin has been associated with premature
aging,
6365
the ability of CS to induce oxidative damage likely
contributes to premature aging.
3.3 |Mechanisms involved beyond PM-
induced skin damage
PM is a complex, heterogeneous mixture of particles, which
vary in size, number, surface areas, concentrations, and chemi-
cal composition. PM particles can be emitted directly from
sources like fossil-fuel combustion as well as generated from
gases through reactions involving other pollutants. PM particles
can be either liquid, solid, or liquid surrounding a solid core
and can be composed of organic chemicals, metals, and soil or
dust particles, as well as nitrates and sulfates, which can be fur-
ther categorized into different particles based on their sizes,
such as PM
10
,PM
2.5
, and ultrafine particles (UFPs). Coarse
particles have a diameter of 2.5 to 10 μm(PM
10
) and can be
generated by farming, mining, and construction.
66
Fine parti-
cleshaveadiameterof2.5μmorless(PM
2.5
)andcanbe
PECORELLI ET AL.5
generated by power plants, oil refineries, fuel combustion, cars,
and wildfires.
66
There are also UFPs with diameters less than
0.1 μm or 100 nm that can be generated by diesel and gasoline
fuel combustion from cars, aircrafts, and ships.
6668
These par-
ticles can differ not only in their size but also in their effects on
human health. A study conducted in 2000 demonstrated that
PM
2.5
particles that are generated by combustion sources were
associated with increased daily mortality.
69
Inhalation of PM
2.5
results in increased plaque deposits in arteries, promoting
development of atherosclerosis as well as increased risk of
heart attacks.
70,71
Most studies on the effects of PM particles on human
health have focused on the negative effects of inhaling these
particles such as asthma, lung cancer, cardiovascular disease,
premature death, and premature delivery and birth defects in
babies. However, epidemiological studies indicate that PM
can promote premature skin aging and exacerbate preexisting
skin diseases.
1
Exposure to PM is associated with progression
of AD in children,
72
and an improvement in air quality
resulted in decreased prevalence and severity of AD.
73
The mechanisms involved in PM-associated skin disor-
ders result from increased oxidative stress due to PM expo-
sure. PMs can move through the skin through hair follicles
or transdermally, generating oxidative stress. PAHs are com-
ponents of UFPs that can be absorbed through the skin and
eventually damage the mitochondria, resulting in intracellu-
lar ROS production.
74
These damaged mitochondria produce
superoxide anions, which can be converted into H
2
O
2
that
can then undergo the Fenton reaction to produce hydroxyl
radicals, resulting in increased ROS and activation of redox-
sensitive transcription factors, such as AP1 and NFκB. In
addition, interactions between PM particles and surfaces can
result in extracellular ROS production, again resulting in the
activation of redox-sensitive transcription factors AP1 and
NFκB. The consequences of oxidative stress result in antiox-
idant depletion, lipid peroxidation, and DNA damage. In
support of this idea, our lab has demonstrated that exposure
to PM particles induces nuclear translocation of NFκB,
increases levels of HNE, and promotes DNA damage in
ex vivo human biopsies.
75
The consequences of increased
oxidative stress in response to PM exposure result in the
exacerbation of preexisting skin diseases and premature skin
aging.
1
3.4 |HNE: the trigger for pollution-induced
skin OxInflammation
HNE derives from the oxidation of ω-6 polyunsaturated fatty
acids (PUFAs), essentially arachidonic and linoleic acid, that
is, the two most represented fatty acids in biomembranes; for a
more in-depth view of HNE production, see.
76
In the context
of pollutants and skin, HNE has been shown to be produced in
the cutaneous tissues after exposure to O
3,
PM, and CS. As
previously mentioned, O
3
immediately interacts with the
PUFAs present in the upper layers of the epithelium, oxidizing
these lipids and forming unstable peroxides that can then lead
to the formation of HNE.
77
The mechanism by which PM is
believed to induce the production of HNE is less direct; the
transition metal constituents of PM (Fe, Zn, Ni, etc.) are
believed to undergo Fenton or Fenton-like reactions, generating
ROS like OH
.78
In addition, as previously mentioned, PAHs,
which are components of PM that are highly lipophilic, can
localize to the mitochondria and promote the generation of
mitochondrial-produced ROS.
79
These increased levels of ROS
can promote the oxidation of ω-6 PUFAs, generating HNE. CS
is also believed to promote the generation of HNE through
increasing ROS as our lab has shown that CS exposure in
keratinocytes increases NAPDH oxidase activity resulting in
increased H
2
O
2
levels.
31
HNE is an unusual compound containing three functional
groups that in many cases act in concert, explaining its high
reactivity (Figure 2). There is, first of all, a conjugated system
consisting of a C=C double bond and a C=O carbonyl group
in HNE. The hydroxyl group at carbon 4 contributes to reactiv-
ity both by polarizing the C=C bond and by facilitating internal
cyclization reactions, such as thioacetal formation.
80,81
HNE is
an amphiphilic molecule; in fact, it is water soluble and also
exhibits strong lipophilic properties. Consequently, HNE tends
to concentrate in biomembranes, where phospholipids, like
phosphatidylethanolamine, and proteins, such as transporters,
ion channels, and receptors, quickly react with HNE. In addi-
tion, since it is a highly electrophilic molecule, it easily reacts
with low molecular weight compounds, such as glutathione,
and at higher concentrations with DNA (Figure 2).
82
Because
of its electrophilic nature, HNE can form adducts with cellular
protein nucleophiles. Indeed, the reactivity of HNE explains its
potential involvement in the modulation of enzyme activity,
signal transduction, and gene expression.
80,81
Besides being a
product of oxidative stress, HNE is also an efficient cell signal-
ing molecule able to modulate the expression of several genes;
therefore, it may influence important cellular functions such as
cell growth, differentiation, and apoptosis. An increasing
amount of literature indicates that HNE, depending on the con-
centrations, can potently activate stress response mechanisms,
such as mitogen-activated protein kinases (MAPKs), detoxifi-
cation mechanisms, and inflammatory responses, contributing
to cell survival against cytotoxic stress. Furthermore, HNE
may modulate redox-sensitive transcription factors such as
nuclear factor-kappa B (NFκB), activator protein-1 (AP1), and
nuclear factor (erythroid-derived 2)-like 2 (Nrf2). Moreover, its
proven interaction with a variety of enzymes and kinases vari-
ously involved in cell signaling strongly support its important
role in pathophysiology as a cell signaling messenger
80,81
(Figure 2).
6PECORELLI ET AL.
4|HNE: METABOLISM, TOXICITY,
AND PROTEIN ADDUCTS
Once formed, under physiological conditions, HNE is rapidly
degraded in mammalian cells by multiple enzymatic pathways.
The best characterized of these enzymes include the glutathione-
S-transferases (GSTs), aldehyde dehydrogenase, and alcohol
dehydrogenase. GSTs catalyze the conjugation of reduced gluta-
thione (GSH) to HNE via Michael addition at the C-3 carbon,
thereby preventing further nucleophilic addition to this toxic
compound. Aldehyde dehydrogenase catalyzes the oxidation of
HNE to the innocuous 4-hydroxy-2-nonenoic acid, while alco-
hol dehydrogenase catalyzes reduction of the terminal aldehyde
to its alcohol, yielding the unreactive metabolite 1,4-dihydroxy-
2-nonene (DHN). Another enzyme involved in the metabolism
of HNE is aldose reductase, a member of the aldoketo reduc-
tase superfamily. This enzyme has been shown to catalyze the
reduction of the GSH conjugate of HNE, leading to DHN
GSH.
80,81
The half-life of HNE has been studied in several cell types,
in subcellular organelles, and even in whole organisms. Liver
tissue generally has the highest capacity to metabolize HNE,
while in other cells, the metabolism of HNE is not so fast, but
still very efficient.
80,81
Usually, HNE, even at very high lipid
peroxidation rates, cannot accumulate in an unlimited manner.
However, compared with other oxidants, such as most types
of ROS, HNE is chemically better suited for its role as a sig-
naling molecule because of its longer half-life and thus greater
range of diffusion and a higher selectivity for reaction with
specific targets. Therefore, despite the fact that humans have
developed several enzymatic systems to rapidly detoxify
HNE molecules, HNE can escape detoxifying processes and
migrate from the site of origin to other intracellular sites,
reacting rapidly with biological macromolecules, especially
proteins to form HNE protein adducts (PAs).
83
HNE PAs are physiological constituents of mammalian
organisms. They are easily detectable in peripheral blood,
where they primarily involve albumin, transferrin, and immu-
noglobulins, and also proteins related to blood coagulation,
lipid transport, blood pressure regulation, and protease inhibi-
tion.
80,81
Nevertheless, as proteins play an important role in
the normal structure and function of cells, oxidative modifica-
tions promoted by increased HNE levels may greatly alter their
structure. These protein alterations may subsequently lead to
loss of normal physiological cell functions and/or may lead to
abnormal function of the cell and eventually to cell death. For
instance, HNE can modify mitochondrial proteins such as
cytochrome c, impairing mitochondrial metabolism.
84
HNE
PAs also contribute to the pool of damaged enzymes, which
increases during aging and in several pathological states.
85
As previously mentioned, HNE can regulate a variety of
normal cell processes including cell growth, differentiation,
FIGURE 2 Schematic
representation of adduct formation by
HNE. (a) 4-Hydroxynonenal (HNE)
contains three functional groups: a C=C
double bond, a C=O carbonyl group, and
a hydroxyl group at carbon 4. All of these
functional groups contribute to its
reactivity. (b,c) HNE can form HNE
protein adducts through Schiff base
addition to lysines and Michael addition to
lysines, histidines, and cysteines.
Specifically, HNE can form adducts with
glutathione through cysteines present in
this protein. (d) HNE can also damage
DNA by forming adducts with
nitrogenous bases (i.e., deoxyguanosine).
In red, we have indicated where bonds
form between HNE and its partner
PECORELLI ET AL.7
and apoptosis. This is likely because HNE can activate a
variety of signal transduction pathways including the Erk
pathway, p38MAPK, c-Jun N-terminal kinases (JNK) path-
way, and epidermal growth factor receptor (EGFR) path-
way.
86
In addition, it can upregulate the extrinsic and
intrinsic apoptotic pathways. Moreover, it can regulate the
activity of critical transcription factors involved in OS
responses such as Nrf2 and peroxisome-proliferator-
activated receptors (PPARs). For instance, HNE can induce
Nrf2 activation by modifying its inhibitor Keap1, releasing
Nrf2 for nuclear export.
87
Moreover, it can enhance the
DNA-binding activity of AP1 and both negatively and posi-
tively regulate NFκB.
86,88
Because of these findings, it is
believed that HNE can be causally involved in many of the
pathophysiological effects associated with oxidative stress in
cells and tissues,
89
especially for the skin due to its rich con-
centration of omega-6 fatty acids. Most studies assessing
HNE production in the skin have primarily focused on
assessing the levels of HNE as a marker of oxidative stress;
therefore, the role of HNE in skin biology remains largely
unexplored.
5|PATHOLOGICAL EFFECTS OF
POLLUTION-INDUCED HNE IN
THE SKIN
Indeed, the presence of HNE adducts in the skin after pollu-
tion exposure has been well documented and in several cases
linked to skin aging, making this marker a possible common
mediator of skin oxidative damage. For instance, HNE levels
were increased after O
3
exposure in both 2D and 3D
models
90,91
and in human skin biopsies after 5 days of O
3
exposure.
41
These results are in line with previous animal
work, wherein hairless mice exposed to O
3
exhibited a clear
increase of HNE PAs in the epidermis.
37
Interestingly, cutane-
ous HNE levels were also significantly higher after O
3
expo-
sure in old animals, compared to young animals, in a wound
healing study, suggesting their role in delaying cutaneous
wound closure in aged mice,
63
possibly via the aberrant acti-
vation of metalloproteinases (MMPs).
64
O
3
is not the only
pollutant that induces the formation of cutaneous HNE in the
skin; indeed, similar effects have been observed after both CS
and PM exposure. Our group was able to demonstrate the for-
mation of HNE PAs in both keratinocytes and reconstituted
human epidermis tissues (RHEs) after CS and PM expo-
sure.
75,92
The formation of HNE PAs, as mentioned before,
leads to the covalent modification of proteins, which can be
subsequently ubiquitinated and degraded. Therefore, a conse-
quence of HNE adduct formation is the loss of important cel-
lular proteins, such as SRB1,
31
which we observed in both
keratinocytes and sebocytes.
30,31
In a very recent work by
Verdin et al. (2019), the ability of UFPs to increase HNE
levels in RHEs was also observed, suggesting a role of HNE
in regulating skin differentiation and cornification.
93
In the skin, we believe that the physiological effects of high
levels of HNE in response to pollutant-induced oxidative stress
results in the development/exacerbation of premature aging,
psoriasis, and AD. Several studies have demonstrated a role for
HNE in skin aging
94,95
and in skin color.
96
In addition, HNE
PAs have been detected in photodamaged skin elastosis.
97
The
FIGURE 3 4-Hydroxynonenal (HNE) as a pollutant-induced signaling mediator. Exposure of the skin to ozone, cigarette smoke, and
particulate matter induces lipid peroxidation and subsequent production of HNE, resulting in the Michael addition of HNE to protein products such
as cytochrome c, which results in ROS production via mitochondria, and Keap1, which releases NrF2 from sequestration. In addition, HNE activates
NAPDH oxidase, again resulting in the generation of ROS and oxidative stress, which can activate NFκB and AhR. HNE can also activate MAPK
pathways, ultimately resulting in the activation of HSPs and transcription factors NFB and AP1. Moreover, modification of cytochrome cby this
signaling mediator can induce caspase activation and PARP1 cleavage. ROS, reactive oxygen species
8PECORELLI ET AL.
effects of HNE in skin aging, associated with pollutant expo-
sure, could be due to its ability to degrade collagen due to alter-
ing MMP levels, which are transcriptionally regulated by
NFκB.
98
A general review of the role of HNE in aging can be
found here.
99
Increased HNE levels have been also detected in
skin samples from psoriatic patients as well as in the cases of
AD.
100,101
Since inflammation is the primary cause of both of
these skin conditions, it is likely that the ability of HNE to reg-
ulate the activity of the key proinflammatory mediator NFκB
contributes to these conditions.
6|CONCLUSION
Cutaneous exposure to pollutants is associated with the dev-
elopment/exacerbation of premature aging, psoriasis, and
AD conditions, and these effects are believed to be mediated
by pollutant-induced oxidative stress. Since oxidative stress
results in the production of HNE, it is no surprise that pollut-
ant exposure results in increased levels of HNE in the epithe-
lium. However, HNE is not just a marker of oxidative stress
but also an important signaling mediator that plays its roles
in a variety of cellular pathways including apoptosis, differ-
entiation, cell growth, and migration through the formation
of HNE adducts as well as modulation of signal transduction
pathways. Thus, being able to prevent cutaneous HNE for-
mation could be a possible innovative cosmeceutical
approach for future topical applications targeting the afore-
mentioned skin conditions (Figure 3).
ACKNOWLEDGMENTS
GV and RP thank the CITYCARE project, funding from the
European Unions Horizon 2020 research and the innovation
program under the Marie Sklodowska-Curie grant agreement
No 765602.
CONFLICT OF INTEREST
The authors declare no potential conflict of interest.
ORCID
Giuseppe Valacchi https://orcid.org/0000-0002-8792-
0947
REFERENCES
1. Vierkotter A. Environmental pollution and skin aging. Hautarzt.
2011;62:577578, 580.
2. Krutmann J, Liu W, Li L, et al. Pollution and skin: From epide-
miological and mechanistic studies to clinical implications.
J Dermatol Sci. 2014;76(3):163168.
3. Mancebo SE, Wang SQ. Recognizing the impact of ambient air
pollution on skin health. J Eur Acad Dermatol Venereol. 2015;29
(12):23262332.
4. Puri P, Nandar SK, Kathuria S, Ramesh V. Effects of air pollu-
tion on the skin: A review. Indian J Dermatol Venereol Leprol.
2017;83(4):415423.
5. Koohgoli R, Hudson L, Naidoo K, Wilkinson S, Chavan B,
Birch-Machin MA. Bad air gets under your skin. Exp Dermatol.
2017;26(5):384387.
6. Goldsmith LA. Skin effects of air pollution. Otolaryngol Head
Neck Surg. 1996;114(2):217219.
7. Wu CC, Bao LJ, Tao S, Zeng EY. Dermal uptake from airborne
organics as an important route of human exposure to E-waste com-
bustion fumes. Environ Sci Technol. 2016;50(13):65996605.
8. Weschler CJ, Bekö G, Koch HM, et al. Transdermal uptake of
diethyl phthalate and di(n-butyl) phthalate directly from air:
Experimental verification. Environ Health Perspect. 2015;123
(10):928934.
9. Morrison GC, Weschler CJ, Bekö G, et al. Role of clothing in
both accelerating and impeding dermal absorption of airborne
SVOCs. J Expo Sci Environ Epidemiol. 2016;26(1):113118.
10. Morrison GC, Andersen HV, Gunnarsen L, Varol D, Uhde E,
Kolarik B. Partitioning of PCBs from air to clothing materials in
a Danish apartment. Indoor Air. 2018;28(1):188197.
11. Beko G, Morrison G, Weschler CJ, et al. Measurements of der-
mal uptake of nicotine directly from air and clothing. Indoor Air.
2017;27(2):427433.
12. Xu F, Yan S, Wu M, et al. Ambient ozone pollution as a risk fac-
tor for skin disorders. Br J Dermatol. 2011;165(1):224225.
13. Kousha T, Valacchi G. The air quality health index and emer-
gency department visits for urticaria in Windsor, Canada.
J Toxicol Environ Health A. 2015;78(8):524533.
14. Leung DY. New insights into atopic dermatitis: Role of skin bar-
rier and immune dysregulation. Allergol Int. 2013;62(2):151161.
15. Tsakok T, Woolf R, Smith CH, Weidinger S, Flohr C. Atopic der-
matitis: The skin barrier and beyond. Br J Dermatol. 2019;180(3):
464474.
16. Elias PM, Steinhoff M. Outside-to-inside(and now back to
outside) pathogenic mechanisms in atopic dermatitis. J Invest
Dermatol. 2008;128(5):10671070.
17. Brandt EB, Sivaprasad U. Th2 cytokines and atopic dermatitis.
J Clin Cell Immunol. 2011;2(3):125.
18. Penard-Morand C, Raherison C, Charpin D, et al. Long-term
exposure to close-proximity air pollution and asthma and aller-
gies in urban children. Eur Respir J. 2010;36(1):3340.
19. Morgenstern V, Zutavern A, Cyrys J, et al. Atopic diseases, aller-
gic sensitization, and exposure to traffic-related air pollution in
children. Am J Respir Crit Care Med. 2008;177(12):13311337.
20. Lee JY, Lamichhane DK, Lee M, et al. Preventive effect of resi-
dential green space on infantile atopic dermatitis associated with
prenatal air pollution exposure. Int J Environ Res Public Health.
2018;15(1):111.
21. Yi O, Kwon HJ, Kim H, et al. Effect of environmental tobacco
smoke on atopic dermatitis among children in Korea. Environ
Res. 2012;113:4045.
22. Shinohara M, Matsumoto K. Fetal tobacco smoke exposure in
the third trimester of pregnancy is associated with atopic
eczema/dermatitis syndrome in infancy. Pediatr Allergy Immunol
Pulmonol. 2017;30(3):155162.
PECORELLI ET AL.9
23. Lee CH, Chuang HY, Hong CH, et al. Lifetime exposure to ciga-
rette smoking and the development of adult-onset atopic dermati-
tis. Br J Dermatol. 2011;164(3):483489.
24. van Voorhis M, Knopp S, Julliard W, et al. Exposure to atmo-
spheric particulate matter enhances Th17 polarization through the
aryl hydrocarbon receptor. PLoS One. 2013;8(12):e82545.
25. Tsuji G, Takahara M, Uchi H, et al. An environmental contaminant,
benzo(a)pyrene, induces oxidative stress-mediated interleukin-8 pro-
duction in human keratinocytes via the aryl hydrocarbon receptor
signaling pathway. J Dermatol Sci. 2011;62(1):4249.
26. Afaq F, Zaid MA, Pelle E, et al. Aryl hydrocarbon receptor is an
ozone sensor in human skin. J Invest Dermatol. 2009;129(10):
23962403.
27. Cella M, Colonna M. Aryl hydrocarbon receptor: Linking envi-
ronment to immunity. Semin Immunol. 2015;27(5):310314.
28. Lee EJ, Han KD, Han JH, Lee JH. Smoking and risk of psoriasis:
A nationwide cohort study. J Am Acad Dermatol. 2017;77(3):
573575.
29. Yang YS, Lim HK, Hong KK, et al. Cigarette smoke-induced
interleukin-1 alpha may be involved in the pathogenesis of adult
acne. Ann Dermatol. 2014;26(1):1116.
30. Crivellari I, Sticozzi C, Belmonte G, et al. SRB1 as a new redox
target of cigarette smoke in human sebocytes. Free Radic Biol
Med. 2017;102:4756.
31. Sticozzi C, Belmonte G, Pecorelli A, et al. Cigarette smoke
affects keratinocytes SRB1 expression and localization via H2O2
production and HNE protein adducts formation. PLoS One.
2012;7(3):e33592.
32. Sticozzi C, Pecorelli A, Romani A, et al. Tropospheric ozone
affects SRB1 levels via oxidative post-translational modifications
in lung cells. Free Radic Biol Med. 2018;126:287295.
33. Muresan XM, Sticozzi C, Belmonte G, et al. SR-B1 involvement
in keratinocytes in vitro wound closure. Arch Biochem Biophys.
2018;658:16.
34. Roberts WE. Pollution as a risk factor for the development of
melasma and other skin disorders of facial hyperpigmentation is
there a case to be made? J Drugs Dermatol. 2015;14(4):337341.
35. Kim KE, Cho D, Park HJ. Air pollution and skin diseases:
Adverse effects of airborne particulate matter on various skin dis-
eases. Life Sci. 2016;152:126134.
36. Valacchi G, Virgili F, Cervellati C, Pecorelli A. OxInflammation:
From subclinical condition to pathological biomarker. Front Phy-
siol. 2018;9:858.
37. Valacchi G, van der Vliet A, Schock BC, et al. Ozone exposure
activates oxidative stress responses in murine skin. Toxicology.
2002;179(12):163170.
38. Valacchi G, Pagnin E, Corbacho AM, et al. In vivo ozone expo-
sure induces antioxidant/stress-related responses in murine lung
and skin. Free Radic Biol Med. 2004;36(5):673681.
39. Packer L, Valacchi G. Antioxidants and the response of skin to
oxidative stress: Vitamin E as a key indicator. Skin Pharmacol
Appl Skin Physiol. 2002;15(5):282290.
40. Valacchi G, Pagnin E, Okamoto T, et al. Induction of stress pro-
teins and MMP-9 by 0.8 ppm of ozone in murine skin. Biochem
Biophys Res Commun. 2003;305(3):741746.
41. Valacchi G, Pecorelli A, Belmonte G, et al. Protective effects of
topical vitamin C compound mixtures against ozone-induced dam-
age in human skin. J Invest Dermatol. 2017;137(6):13731375.
42. Pryor WA. Mechanisms of radical formation from reactions of
ozone with target molecules in the lung. Free Radic Biol Med.
1994;17(5):451465.
43. Valavanidis A, Vlachogianni T, Fiotakis K. Tobacco smoke:
Involvement of reactive oxygen species and stable free radicals in
mechanisms of oxidative damage, carcinogenesis and synergistic
effects with other respirable particles. Int J Environ Res Public
Health. 2009;6(2):445462.
44. Church DF, Pryor WA. Free-radical chemistry of cigarette smoke
and its toxicological implications. Environ Health Perspect.
1985;64:111126.
45. Schick S, Glantz S. Philip Morris toxicological experiments with
fresh sidestream smoke: More toxic than mainstream smoke. Tob
Control. 2005;14(6):396404.
46. Freiman A, Bird G, Metelitsa AI, Barankin B, Lauzon GJ. Cuta-
neous effects of smoking. J Cutan Med Surg. 2004;8(6):415423.
47. Morita A. Tobacco smoke causes premature skin aging. J Dermatol
Sci. 2007;48(3):169175.
48. Grether-Beck S, Wlaschek M, Krutmann J, Scharffetter-
Kochanek K. Photodamage and photoagingPrevention and
treatment. J Dtsch Dermatol Ges. 2005;3(Suppl 2):S19S25.
49. Fisher GJ, Talwar HS, Lin J, Voorhees JJ. Molecular mechanisms
of photoaging in human skin in vivo and their prevention by all-
trans retinoic acid. Photochem Photobiol. 1999;69(2):154157.
50. Model D. Smoker's face: An underrated clinical sign? Br Med J
(Clin Res Ed). 1985;291(6511):17601762.
51. Daniell HW. Smoker's wrinkles. A study in the epidemiology of
"crow's feet". Ann Intern Med. 1971;75(6):873880.
52. Morita A, Torii K, Maeda A, Yamaguchi Y. Molecular basis of
tobacco smoke-induced premature skin aging. J Investig
Dermatol Symp Proc. 2009;14(1):5355.
53. Yin L, Morita A, Tsuji T. Alterations of extracellular matrix
induced by tobacco smoke extract. Arch Dermatol Res. 2000;292
(4):188194.
54. Carver LA, LaPres JJ, Jain S, Dunham EE, Bradfield CA. Char-
acterization of the ah receptor-associated protein, ARA9. J Biol
Chem. 1998;273(50):3358033587.
55. Kazlauskas A, Poellinger L, Pongratz I. The immunophilin-like
protein XAP2 regulates ubiquitination and subcellular localiza-
tion of the dioxin receptor. J Biol Chem. 2000;275(52):
4131741324.
56. Reyes H, Reisz-Porszasz S, Hankinson O. Identification of the
Ah receptor nuclear translocator protein (Arnt) as a component of
the DNA binding form of the Ah receptor. Science. 1992;256
(5060):11931195.
57. Watson AJ, Hankinson O. Dioxin-and ah receptor-dependent pro-
tein binding to xenobiotic responsive elements and G-rich DNA
studied by in vivo footprinting. J Biol Chem. 1992;267(10):
68746878.
58. Fujii-Kuriyama Y, Imataka H, Sogawa K, Yasumoto K,
Kikuchi Y. Regulation of CYP1A1 expression. FASEB J. 1992;6
(2):706710.
59. Sutter TR, Guzman K, Dold KM, Greenlee WF. Targets for
dioxin: Genes for plasminogen activator inhibitor-2 and
interleukin-1 beta. Science. 1991;254(5030):415418.
60. Yin H, Li Y, Sutter TR. Dioxin-enhanced expression of
interleukin-1 beta in human epidermal keratinocytes: Potential
role in the modulation of immune and inflammatory responses.
Exp Clin Immunogenet. 1994;11(23):128135.
10 PECORELLI ET AL.
61. Ono Y, Torii K, Fritsche E, et al. Role of the aryl hydrocarbon
receptor in tobacco smoke extract-induced matrix metalloproteinase-
1 expression. Exp Dermatol. 2013;22(5):349353.
62. Yun MR, Park HM, Seo KW, Lee SJ, Im DS, Kim CD.
5-Lipoxygenase plays an essential role in 4-HNE-enhanced ROS
production in murine macrophages via activation of NADPH oxi-
dase. Free Radic Res. 2010;44(7):742750.
63. Lim Y, Phung AD, Corbacho AM, et al. Modulation of cutaneous
wound healing by ozone: Differences between young and aged
mice. Toxicol Lett. 2006;160(2):127134.
64. Fortino V, Maioli E, Torricelli C, Davis P, Valacchi G. Cutane-
ous MMPs are differently modulated by environmental stressors
in old and young mice. Toxicol Lett. 2007;173(2):7379.
65. Stadtman ER. Role of oxidant species in aging. Curr Med Chem.
2004;11(9):11051112.
66. Araujo JA, Nel AE. Particulate matter and atherosclerosis: Role
of particle size, composition and oxidative stress. Part Fibre
Toxicol. 2009;6:24.
67. Franck U, Odeh S, Wiedensohler A, Wehner B, Herbarth O. The
effect of particle size on cardiovascular disordersthe smaller the
worse. Sci Total Environ. 2011;409(20):42174221.
68. Heal MR, Kumar P, Harrison RM. Particles, air quality, policy
and health. Chem Soc Rev. 2012;41(19):66066630.
69. Laden F, Neas LM, Dockery DW, Schwartz J. Association of fine
particulate matter from different sources with daily mortality in
six U.S. cities. Environ Health Perspect. 2000;108(10):941947.
70. Pope CA 3rd, Burnett RT, Thun MJ, et al. Lung cancer, cardio-
pulmonary mortality, and long-term exposure to fine particulate
air pollution. JAMA. 2002;287(9):11321141.
71. Cesaroni G, Forastiere F, Stafoggia M, et al. Long term exposure
to ambient air pollution and incidence of acute coronary events:
Prospective cohort study and meta-analysis in 11 European
cohorts from the ESCAPE project. BMJ. 2014;348:f7412.
72. Song S, Lee K, Lee YM, et al. Acute health effects of urban fine
and ultrafine particles on children with atopic dermatitis. Environ
Res. 2011;111(3):394399.
73. Kim HO, Kim JH, Cho SI, et al. Improvement of atopic dermati-
tis severity after reducing indoor air pollutants. Ann Dermatol.
2013;25(3):292297.
74. Li N, Sioutas C, Cho A, et al. Ultrafine particulate pollutants
induce oxidative stress and mitochondrial damage. Environ
Health Perspect. 2003;111(4):455460.
75. Magnani ND, Muresan XM, Belmonte G, et al. Skin damage
mechanisms related to airborne particulate matter exposure.
Toxicol Sci. 2016;149(1):227236.
76. Csala M, Kardon T, Legeza B, et al. On the role of
4-hydroxynonenal in health and disease. Biochim Biophys Acta.
2015;1852(5):826838.
77. Zeng J, Lu J. Mechanisms of action involved in ozone-therapy in
skin diseases. Int Immunopharmacol. 2018;56:235241.
78. Chen LC, Lippmann M. Effects of metals within ambient air partic-
ulate matter (PM) on human health. Inhal Toxicol. 2009;21(1):131.
79. Li N, Xia T, Nel AE. The role of oxidative stress in ambient par-
ticulate matter-induced lung diseases and its implications in the
toxicity of engineered nanoparticles. Free Radic Biol Med. 2008;
44(9):16891699.
80. Poli G, Biasi F, Leonarduzzi G. 4-Hydroxynonenal-protein
adducts: A reliable biomarker of lipid oxidation in liver diseases.
Mol Aspects Med. 2008;29(12):6771.
81. Poli G, Schaur RJ, Siems WG, Leonarduzzi G. 4-hydroxynonenal:
A membrane lipid oxidation product of medicinal interest. Med
Res Rev. 2008;28(4):569631.
82. Esterbauer H, Schaur RJ, Zollner H. Chemistry and biochemistry
of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free
Radic Biol Med. 1991;11(1):81128.
83. Petersen DR, Doorn JA. Reactions of 4-hydroxynonenal with pro-
teins and cellular targets. Free Radic Biol Med. 2004;37(7):937945.
84. Isom AL, Barnes S, Wilson L, Kirk M, Coward L, Darley-
Usmar V. Modification of cytochrome c by 4-hydroxy-2-nonenal:
Evidence for histidine, lysine, and arginine-aldehyde adducts.
J Am Soc Mass Spectrom. 2004;15(8):11361147.
85. Uchida K. 4-Hydroxy-2-nonenal: A product and mediator of oxi-
dative stress. Prog Lipid Res. 2003;42(4):318343.
86. Iles KE, Dickinson DA, Wigley AF, Welty NE, Blank V,
Forman HJ. HNE increases HO-1 through activation of the ERK
pathway in pulmonary epithelial cells. Free Radic Biol Med.
2005;39(3):355364.
87. Dinkova-Kostova AT, Holtzclaw WD, Cole RN, et al. Direct evi-
dence that sulfhydryl groups of Keap1 are the sensors regulating
induction of phase 2 enzymes that protect against carcinogens and
oxidants. Proc Natl Acad Sci USA. 2002;99(18):1190811913.
88. Wang S, Kotamraju S, Konorev E, Kalivendi S, Joseph J,
Kalyanaraman B. Activation of nuclear factor-kappaB during
doxorubicin-induced apoptosis in endothelial cells and myocytes
is pro-apoptotic: The role of hydrogen peroxide. Biochem J.
2002;367(Pt 3:729740.
89. Dubinina EE, Dadali VA. Role of 4-hydroxy-trans-2-nonenal in
cell functions. Biochemistry (Mosc.). 2010;75(9):10691087.
90. Valacchi G, Muresan XM, Sticozzi C, et al. Ozone-induced dam-
age in 3D-skin Model is prevented by topical vitamin C and vita-
min E compound mixtures application. J Dermatol Sci. 2016;82
(3):209212.
91. Valacchi G, Sticozzi C, Belmonte G, et al. Vitamin C compound
mixtures prevent ozone-induced oxidative damage in human
keratinocytes as initial assessment of pollution protection. PLoS
One. 2015;10(8):e0131097.
92. Romani A, Cervellati C, Muresan XM, et al. Keratinocytes oxi-
dative damage mechanisms related to airbone particle matter
exposure. Mech Ageing Dev. 2018;172:8695.
93. Verdin A, Cazier F, Fitoussi R, et al. An in vitro model to evalu-
ate the impact of environmental fine particles (PM0.3-2.5) on
skin damage. Toxicol Lett. 2019;305:94102.
94. Poot M, Verkerk A, Koster JF, Esterbauer H, Jongkind JF. Influ-
ence of cumene hydroperoxide and 4-hydroxynonenal on the glu-
tathione metabolism during in vitro ageing of human skin
fibroblasts. Eur J Biochem. 1987;162(2):287291.
95. Jorgensen P, Milkovic L, Zarkovic N, Waeg G, Rattan SI. Lipid
peroxidation-derived 4-hydroxynonenal-modified proteins accu-
mulate in human facial skin fibroblasts during ageing in vitro.
Biogerontology. 2014;15(1):105110.
96. Ogura Y, Kuwahara T, Akiyama M, et al. Dermal carbonyl modi-
fication is related to the yellowish color change of photo-aged
Japanese facial skin. J Dermatol Sci. 2011;64(1):4552.
97. Tanaka N, Tajima S, Ishibashi A, Uchida K, Shigematsu T.
Immunohistochemical detection of lipid peroxidation products,
protein-bound acrolein and 4-hydroxynonenal protein adducts, in
actinic elastosis of photodamaged skin. Arch Dermatol Res.
2001;293(7):363367.
PECORELLI ET AL.11
98. Bond M, Chase AJ, Baker AH, Newby AC. Inhibition of tran-
scription factor NF-kappaB reduces matrix metalloproteinase-1,
3 and 9 production by vascular smooth muscle cells. Cardi-
ovasc. Res. 2001;50(3):556565.
99. Zhang H, Forman HJ. 4-hydroxynonenal-mediated signaling and
aging. Free Radic Biol Med. 2017;111:219225.
100. Gegotek A, Domingues P, Wro
nski A, Ambrożewicz E,
Skrzydlewska E. The proteomic profile of keratinocytes and lym-
phocytes in psoriatic patients. Proteomics Clin Appl. 2019;
e1800119.
101. Niwa Y, Sumi H, Kawahira K, Terashima T, Nakamura T,
Akamatsu H. Protein oxidative damage in the stratum corneum:
Evidence for a link between environmental oxidants and the
changing prevalence and nature of atopic dermatitis in Japan. Br
J Dermatol. 2003;149(2):248254.
How to cite this article: Pecorelli A, Woodby B,
Prieux R, Valacchi G. Involvement of 4-hydroxy-2-
nonenal in pollution-induced skin damage.
BioFactors. 2019;112. https://doi.org/10.1002/
biof.1513
12 PECORELLI ET AL.
... Indeed, exposure to environmental pollutants is known to cause oxidative and inflammatory reactions within the human skin that can ultimately result in skin structural damage and premature aging [64]. ROS induced by air pollutants can promote lipid peroxidative events that culminate in the production of secondary mediators, such as the reactive aldehyde 4HNE, that can propagate cutaneous oxidative damage throughout the skin layers [65]. 4HNE can affect skin proteins' functionality by forming adducts that cause such proteins to lose their activity and be degraded [65]. ...
... ROS induced by air pollutants can promote lipid peroxidative events that culminate in the production of secondary mediators, such as the reactive aldehyde 4HNE, that can propagate cutaneous oxidative damage throughout the skin layers [65]. 4HNE can affect skin proteins' functionality by forming adducts that cause such proteins to lose their activity and be degraded [65]. As a matter of fact, the ex vivo studies demonstrated that the human skin exposed to both SOL-PIP and SOL-CUR failed to prevent oxidative damage from DEE, aligning with previous findings of SOL-PIP's lack of antioxidant activity and SOL-CUR's poor permeability. ...
Article
Full-text available
Diesel particulate matter is one of the most dangerous environmental stressors affecting human health. Many plant-derived compounds with antioxidant and anti-inflammatory properties have been proposed to protect the skin from pollution damage. Curcumin (CUR) has a plethora of pharmacological activities, including anticancer, antimicrobial, anti-inflammatory and antioxidant. However, it has low bioavailability due to its difficult absorption and rapid metabolism and elimination. CUR encapsulation in nanotechnological systems and its combination with biopotentiators such as piperine (PIP) can improve its pharmacokinetics, stability and activity. In this study, ethosomes (ETs) were investigated for CUR and PIP delivery to protect the skin from damage induced by diesel particulate matter. ETs were produced by different strategies and characterized for their size distribution by photon correlation spectroscopy, for their morphology by transmission electron microscopy, and for their drug encapsulation efficiency by high-performance liquid chromatography. Franz cells enabled us to evaluate in vitro the drug diffusion from ETs. The results highlighted that ETs can promote the skin permeation of curcumin. The studies carried out on their antioxidant activity demonstrated an increase in the antioxidant power of CUR using a combination of CUR and PIP separately loaded in ETs, suggesting their possible application for the prevention of skin damage due to exogenous stressors. Ex vivo studies on human skin explants have shown the suitability of drug-loaded ETs to prevent the structural damage to the skin induced by diesel engine exhaust exposure.
... CS is a complex aerosol composed of a mixture of more than 4700 chemical substances, among which 1014 are low-molecular-weight carbon-and oxygen-centered radicals [12,13] that can participate in oxidative events [14]. In particular, the main cause of oxidative stress reactions linked to CS is represented by the production of reactive aldehydes during lipid peroxidation events (4-hydroxy-2,3-nonenal and malondialdehyde) [15], or due to the presence of α,β-unsaturated aldehyde species (acrolein and crotonaldehyde) in the CS composition [16]. A similar mechanism of action is ascribed to O 3 , which, although too reactive to penetrate the skin, can directly interact with skin biomolecules, such as lipids, proteins, or DNA, leading to the production of ROS and other nonradical species such as aldehydes (4-hydroxy-nonenal, 4HNE), which can promote the alteration of its structure and function [17,18]. ...
... First, our results showed that exposure to air pollutants, particularly to MP and UV, promoted increased levels of 4HNE (Figure 1a), one of the most reactive products of lipid peroxidation. Since air pollutants can interact with the lipids present within the stratum corneum, leading to the production of ROS and consequent lipid peroxidation, 4HNE represents an important oxidative stress marker in skin conditions [15,42]. Moreover, 4HNE is involved in the regulation of several inflammatory and oxidative stress pathways, including the activation of redox-sensitive transcription factors such as NF-κB and NRF2. ...
Article
Full-text available
The skin is the outermost layer of the body and, therefore, is exposed to a variety of stressors, such as environmental pollutants, known to cause oxinflammatory reactions involved in the exacerbation of several skin conditions. Today, inflammasomes are recognized as important modulators of the cutaneous inflammatory status in response to air pollutants and ultraviolet (UV) light exposure. In this study, human skin explants were exposed to the best-recognized air pollutants, such as microplastics (MP), cigarette smoke (CS), diesel engine exhaust (DEE), ozone (O3), and UV, for 1 or 4 days, to explore how each pollutant can differently modulate markers of cutaneous oxinflammation. Exposure to environmental pollutants caused an altered oxidative stress response, accompanied by increased DNA damage and signs of premature skin aging. The effect of specific pollutants being able to exert different inflammasomes pathways (NLRP1, NLRP3, NLRP6, and NLRC4) was also investigated in terms of scaffold formation and cell pyroptosis. Among all environmental pollutants, O3, MP, and UV represented the main pollutants affecting cutaneous redox homeostasis; of note, the NLRP1 and NLRP6 inflammasomes were the main ones modulated by these outdoor stressors, suggesting their role as possible molecular targets in preventing skin disorders and the inflammaging events associated with environmental pollutant exposure.
... On the other hand, PM impairs intracellular antioxidant response systems, such as the inhibition of intracellular glutathione peroxidase, and decreases the superoxide dismutases, putting cells in a state of oxidative stress (Guo et al., 2022a;Patatian et al., 2021;Wang et al., 2021c). In addition, cell phagocytosis of contaminant particles and activation of AhR by PAH promote intracellular ROS (Furue et al., 2014;Herath et al., 2022;Li et al., 2017a;Pecorelli et al., 2019). Inflammation is a key factor in the development and exacerbation of inflammatory skin diseases. ...
... PM 10 could upregulate the expression of inflammatory cytokines such as interleukin (IL)-1β, IL-4, IL-6, IL-17α, IL-25, IL-31, and thymus stromal lymphopoietin (Kwack et al., 2022a;Kwack et al., 2022d). In another way, PM exposure induces lipid peroxidation (i.e., production of 4-hydroxynonanal) and ROS, thereby stimulating the activation of inflammasomes (Pecorelli et al., 2019). Then inflammasome activation leads to the development/exacerbation of psoriasis, AD, and acne (Ferrara et al., 2020). ...
... 7,8 O 3 interacts with lipids and proteins present in the stratum corneum, resulting in the production of reactive oxygen species (ROS), secondary mediators such as aldehydes (4-HydroxyNonenal, 4-HNE) and cyclooxygenase 2 (COX-2), and in the activation of inflammasome pathways, such as NLR Family Pyrin Domain Containing (NLRP1). [9][10][11][12][13][14][15] This process causes oxidative damage throughout the skin and initiates inflammatory processes. 11,[16][17][18][19][20] A growing set of data demonstrated that different pollutants affect skin expression levels of the transcription factor aryl hydrocarbon receptor (AhR) and that its activation in skin cells parallels that of keratinocyte differentiation and inflammation, resulting in cutaneous damage, skin diseases and premature skin aging. ...
... [9][10][11][12][13][14][15] This process causes oxidative damage throughout the skin and initiates inflammatory processes. 11,[16][17][18][19][20] A growing set of data demonstrated that different pollutants affect skin expression levels of the transcription factor aryl hydrocarbon receptor (AhR) and that its activation in skin cells parallels that of keratinocyte differentiation and inflammation, resulting in cutaneous damage, skin diseases and premature skin aging. [21][22][23] Moreover, O 3 and UV alter the expression of the stratum corneum cornified envelope components such as lipids, proteins (Keratin, Filaggrin, Involucrin), as well as tight junctions. ...
Article
Full-text available
Purpose Exposure of the skin to ultraviolet radiation (UV) or ozone (O3) results in stressed skin, leading to the alteration of the skin physical barrier and defence functions. In this work, the preventive benefit of a dermocosmetic, M89PF, containing Vichy mineralising water, probiotic fractions, antioxidant vitamins and hyaluronic acid, in the alteration of skin physical barrier and skin defence functions after exposure to O3 and UV, alone or combined, was assessed. Methods Untreated and treated (M89PF) skin explants were exposed to O3, to UV rays or to O3+UV. Immunofluorescence was performed for skin barrier, oxidative stress, and inflammatory markers after one and four days of exposure to the pollutants. Results M89PF significantly (p≤0.05) prevented the decrease of the expression level of different skin barrier markers, and significantly (p≤0.05) prevented the induction of OxInflammatory markers and inflammasome components by UV, O3, or both combined. Conclusion M89PF prevents skin barrier damage, as well as oxidative stress and inflammatory markers induced by exposome factors, such as UV, O3, or both combined.
... generate reactive oxygen species (ROS) that may lead to secondary events as DNA damage, protein oxidation and lipid peroxidation in the cutaneous tissue.10 Among the lipid peroxidation byproducts, the aldehyde 4-hydroxynonenal (4HNE) is one of the most abundant and reactive, being able to form covalent adducts with macromolecules, affecting their structure and function and promoting their degradation via the proteasome.31 Our data show that exposure to PM + UV promoted increased levels of 4HNE protein adducts (4HNE PA), as the result of oxidative stress reactions within the cutaneous tissue(Figure 2A), whereas pre-treatment with CF Mix clearly prevented the protein oxidative damage in human skin biopsies by reducing the levels of 4HNE PA of 48.91% respect to PM + UV exposed tissues. ...
Article
Full-text available
Background Exposure to environmental stressors like particulate matter (PM) and ultraviolet radiation (UV) induces cutaneous oxidative stress and inflammation and leads to skin barrier dysfunction and premature aging. Metals like iron or copper are abundant in PM and are known to contribute to reactive oxygen species (ROS) production. Aims Although it has been suggested that topical antioxidants may be able to help in preventing and/or reducing outdoor skin damage, limited clinical evidence under real‐life exposure conditions have been reported. The aim of the present study was to evaluate the ability of a topical serum containing 15% ascorbic acid, 0.5% ferulic acid, and 1% tocopherol (CF Mix) to prevent oxinflammatory skin damage and premature aging induced by PM + UV in a human clinical trial. Methods A 4‐day single‐blinded, clinical study was conducted on the back of 15 females (18–40 years old). During the 4 consecutive days, the back test zones were treated daily with or without the CF Mix, followed by with/without 2 h of PM and 5 min of UV daily exposure. Results Application of the CF Mix prevented PM + UV‐induced skin barrier perturbation (Involucrin and Loricrin), lipid peroxidation (4HNE), inflammatory markers (COX2, NLRP1, and AhR), and MMP9 activation. In addition, CF Mix was able to prevent Type I Collagen loss. Conclusion This is the first human study confirming multipollutant cutaneous damage and suggesting the utility of a daily antioxidant topical application to prevent pollution induced skin damage.
... Within SASP, the 4-hydroxy-2-nonenal (4-HNE) is another molecule associated with inflammatory processes. 4-HNE is a highly reactive product of lipid peroxidation caused by the action of ROS, which can form adducts with cellular proteins and even DNA [148]. Human keratinocytes exposed to O 3 inhibited cell proliferation and stimulated increased oxidative damage, which increased levels of 4-HNE and activated the NF-κB pathway, demonstrating that O 3 is a potential inducer of skin inflammation [99]. ...
Article
Full-text available
Our current understanding of skin cell senescence involves the role of environmental stressors (UV, O3, cigarette smoke, particulate matter, etc.), lifestyle (diet, exercise, etc.) as well as genetic factors (metabolic changes, hormonal, etc.). The common mechanism of action of these stressors is the disturbance of cellular redox balance characterized by increased free radicals and reactive oxygen species (ROS), and when these overload the intrinsic antioxidant defense system, it can lead to an oxidative stress cellular condition. The main redox mechanisms that activate cellular senescence in the skin involve (1) the oxidative damage of telomeres causing their shortening; (2) the oxidation of proteomes and DNA damage; (3) an a in lysosomal mass through the increased activity of resident enzymes such as senescence-associated β-galactosidase (SA-β-gal) as well as other proteins that are products of lysosomal activity; (4) and the increased expression of SASP, in particular pro-inflammatory cytokines transcriptionally regulated by NF-κB. However, the main targets of ROS on the skin are the proteome (oxi-proteome), followed by telomeres, nucleic acids (DNAs), lipids, proteins, and cytoplasmic organelles. As a result, cell cycle arrest pathways, lipid peroxidation, increased lysosomal content and dysfunctional mitochondria, and SASP synthesis occur. Furthermore, oxidative stress in skin cells increases the activity of p16INK4A and p53 as inhibitors of Rb and CDks, which are important for maintaining the cell cycle. p53 also promotes the inactivation of mTOR-mediated autophagic and apoptotic pathways, leading to senescence. However, these markers alone cannot establish the state of cellular senescence, and multiple analyses are encouraged for confirmation. An updated and more comprehensive approach to investigating skin senescence should include further assays of ox-inflammatory molecular pathways that can consolidate the understanding of cutaneous redox senescence.
... The mechanism by which ozone induces cutaneous damage is via the direct interaction and oxidation of polyunsaturated fatty acids (PUFAs) and lipids present in the outermost layer of skin, the stratum corneum (SC), leading to the direct generation of aldehydes (i.e., 4-hydroxynonenal-4HNE) and reactive oxygen species (ROS) including H 2 O 2 . All these mediators are able to further carry on ozone toxicity affecting the integrity and functionality of skin barrier as well as perpetuating the damage in deeper cutaneous layers [11][12][13] . ...
Article
Full-text available
Being the more apparent organ exposed to the outdoor stressors, the effect of pollution on the skin has been widely studied in the last few decades. Although UV light is known as the most aggressive stressor to which our cutaneous tissue is daily exposed, other components of the tropospheric pollution have also shown to affect skin health and functionality. Among them, ozone has been proven to be one of the most toxic due to its high reactivity with the epidermal lipids. Studying the cutaneous effect of pollution in a laboratory setting presents challenges, therefore it becomes critical to employ appropriate and tailored models that aim to answer specific questions. Several skin models are available nowadays: in vitro models (2D cell lines and 3D cutaneous tissues), ex vivo skin explants and in vivo approaches (animals and humans). Although in the last 20 years researchers developed skin models that closely resemble human skin (3D cutaneous tissues), ex vivo skin explants still remain one of the best models to study cutaneous responses. Unfortunately, one important cutaneous property that is not present in the traditional ex vivo human skin explants is the physiological tension, which has been shown to be a cardinal player in skin structure, homeostasis, functional properties and responses to external stimuli. For this reason, in this study, to confirm and further comprehend the harmful mechanism of ozone exposure on the integumentary system, we have performed experiments using the state of art in cutaneous models: the innovative TenSkin™ model in which ex vivo human skin explants are cultured under physiologically relevant tension during the whole experimental procedure. Specifically, we were interested in corroborating previous findings showing that ozone exposure modulates the expression of cutaneous antimicrobial peptides (AMPs). The present work demonstrates that cutaneous exposure to ozone induces AMPs gene and protein levels (CAMP/LL-37, hBD2, hBD3) and that the presence of tension can further modulate their expression. In addition, different responses between tension and non-tension cultured skin were also observed during the evaluation of OxInflammatory markers [cyclooxygenase-2 (COX2), aryl hydrocarbon receptor (AhR), matrix-metallo-proteinase 9 (MMP9) and 4-hydroxy-nonenal (4HNE)]. This current study supports our previous findings confirming the ability of pollution to induce the cutaneous expression of AMPs via redox signaling and corroborates the principle that skin explants are a good and reliable model to study skin responses even though it underlines the need to holistically consider the role of skin tension before extrapolating the data to real life.
... Ozone (O 3 ) is one of the most recognized oxidative air pollutants that can react with skin macromolecules such as lipids, proteins, etc. and induce oxidative stress reactions culminating in the formation of lipid peroxidation products like the reactive aldehyde 4-hydroxy-nonenal (4HNE) [42]. 4HNE can then promote the formation of protein adducts within the skin that can alter the cutaneous homeostasis, favoring an oxidative environment. ...
Article
Full-text available
In this study, we examined and compared two different lipid-based nanosystems (LBNs), namely Transferosomes (TFs) and Monoolein Aqueous Dispersions (MADs), as delivery systems for the topical application of Ferulic Acid (FA), an antioxidant molecule derived from natural sources. Our results, as demonstrated through Franz-cell experiments, indicate that the LBNs produced with poloxamer 188 in their composition create a multilamellar system. This system effectively controls the release of the drug. Nonetheless, we found that the type of non-ionic surfactant can impact the drug release rate. Regarding FA diffusion from the MAD, this showed a lower diffusion rate compared with the TF. In terms of an in vivo application, patch tests revealed that all LBN formulations tested were safe when applied under occlusive conditions for 48 h. Additionally, human skin biopsies were used to determine whether FA-containing formulations could influence skin tissue morphology or provide protection against O3 exposure. Analyses suggest that treatment with TFs composed of poloxamer 188 and MAD formulations might protect against structural skin damage (as observed in hematoxylin/eosin staining) and the development of an oxidative environment (as indicated by 4-hyroxinonenal (4HNE) expression levels) induced by O3 exposure. In contrast, formulations without the active ingredient did not offer protection against the detrimental effects of O3 exposure.Inizio modulo.
Article
Full-text available
The skin is constantly exposed to a variety of environmental threats. Therefore, the influence of environmental factors on skin damage has always been a matter of concern. This study aimed to investigate the cytotoxic effects of different environmental factors, including cooking oil fumes (COFs), haze (PM2.5), and cigarette smoke (CS), on epidermal HaCaT cells and dermal fibroblast (FB) cells. Cell viability, intracellular reactive oxygen species (ROS) generation, inflammatory cytokine levels, and collagen mRNA expression were used as toxicity endpoints. Additionally, the effects of ozone (O3) on cell viability and release of inflammatory cytokines in 3D epidermal cells were also examined. The results showed that the organic extracts of CS, COFs, and PM2.5 significantly inhibited the viability of HaCaT and FB cells at higher exposure concentrations. These extracts also increased intracellular ROS levels in FB cells. Furthermore, they significantly promoted the release of inflammatory cytokines, such as IL-1α and TNF-α, in HaCaT cells and down-regulated the mRNA expression of collagen I, III, IV, and VII in FB cells. Comparatively, SC organic extracts exhibited stronger cytotoxicity to skin cells compared to PM2.5 and COFs. Additionally, O3 at all test concentrations significantly inhibited the viability of 3D epidermal cells in a concentration-dependent manner and markedly increased the levels of TNF-α and IL-1α in 3D epidermal cells. These findings emphasize the potential cytotoxicity of COFs, PM2.5, CS, and O3 to skin cells, which may lead to skin damage; therefore, we should pay attention to these environmental factors and take appropriate measures to protect the skin from their harmful effects.
Article
Full-text available
Exposure to airborne particulate matter (PM) has significant effects on human health mainly leading to cardio-respiratory diseases. However very few data are available regarding the impact of PM on the skin, so to better understand the impact of fine particle (PM 0.3-2.5 ) on both inflammatory response and epidermal structure, we exposed a reconstructed human epidermis (RHE) to several doses of PM collected in Cotonou (Benin, West Africa). After 24 h of exposure, inflammatory response, histological observations, and gene expression related to oxidative stress, antioxidant defense and structural damages were determined. No PM-linked changes in tissue morphology or membrane integrity were observable. PM was however cytotoxic in a dose dependent manner. An inflammatory response appeared as shown by the increase in IL-1α and IL-8 cytokine productions. PM also induced oxidative stress, leading to an increase in 4-HNE immunostaining and to the up-regulation of HMOX1, MT1G and MT1E. Finally, PM had a negative impact on fundamental skin functions such as tissue anchorage, cell differentiation, cornification / skin desquamation and apoptosis. Our data show that airborne fine particles have an adverse effect on skin integrity, most probably leading to accelerated ageing.
Article
Full-text available
Inflammation is a complex systemic response evolved to cope with cellular injury, either due to infectious agents or, in general, with sporadic events challenging tissue integrity and function. Researchers involved in different fields have the tendency to look at the inflammatory response with different angles, according to their specific interest. Established its complexity, one of the most evident features of the inflammatory response is the generation of a pro-oxidative environment due to the production of high fluxes of pro-oxidant species. This production begins locally, close to the sites of tissue damage or infection, but eventually becomes a chronic challenge for the organism, if the inflammatory response is not properly controlled. In this review, we focus on this specific aspect of chronic, low-level sub-clinical inflammatory response. We propose the term “OxInflammation” as a novel operative term describing a permanent pro-oxidative feature that interact, in a positive feed-back manner, to a not yet clinically detectable inflammatory process, leading in a long run (chronically) to a systemic/local damage, as a consequence of the cross talk between inflammatory, and oxidative stress mediators. Therefore, it could be useful to analyze inflammatory markers in pathologies where there is an alteration of the redox homeostasis, although an inflammatory status is not clinically evident.
Article
Full-text available
Few birth cohort studies have examined the role of traffic-related air pollution (TRAP) in the development of infantile atopic dermatitis (AD), but none have investigated the role of preventive factors such as green spaces. The aim of this study was to investigate whether exposure to nitrogen dioxide (NO2) and particulate matter with an aerodynamic diameter of <10 μm (PM10) during pregnancy is associated with increased risk of development of AD in 6-month-old children and also to examine how this association changes with residential green space. This study used prospective data from 659 participants of the Mothers and Children’s Environmental Health study. Subjects were geocoded to their residential addresses and matched with air pollution data modeled using land-use regression. Information on infantile AD was obtained by using a questionnaire administered to the parents or guardians of the children. The association between infantile AD and exposure to NO2 and PM10 was determined using logistic regression models. We assessed the effects of residential green spaces using stratified analyses and by entering product terms into the logistic regression models. The risk of infantile AD significantly increased with an increase in air pollution exposure during the first trimester of pregnancy. The adjusted odds ratio (OR) and 95% confidence interval (CI) were 1.219 (1.023–1.452) per 10 μg/m3 increase in PM10 and 1.353 (1.027–1.782) per 10 ppb increase in NO2. An increase in the green space within 200 m of residence was associated with a decreased risk of AD (OR = 0.996, 95% CI: 0.993–0.999). The stratified analysis of residential green space revealed stronger associations between infantile AD and PM10 and NO2 exposure during the first trimester in the areas in the lower tertiles of green space. This study indicated that exposure to TRAP during the first trimester of pregnancy is associated with infantile AD. Less residential green space may intensify the association between TRAP exposure and infantile AD.
Article
The skin is a target organ for pollution and also allows the penetration of exogenous agents into the body. About 700,000 new cases of skin cancer were diagnosed in 1993, and 9100 people died of cancer; 76% of the deaths were due to melanoma. Skin cancers are most closely associated with exposure to UVB (290 to 320 nm) irradiation. For every 1% decrease in ozone there is a 2% increase in UVB irradiance, and therefore a 2% increase in skin cancer is predicted. Therefore the atmospheric pollution by ozone-depleting chemicals is a major concern to dermatologists. In addition to being a target organ and site of neoplasms and contact allergens, the skin is the site of significant absorption of environmental pollutants. In the case of chloroform, the percutaneous absorption is equivalent to the respiratory uptake, emphasizing how important it is to recognize skin absorption in toxicologic exposures.
Article
Background Cigarette smoking is the single biggest preventable cause of death and disability in developed countries and is a significant public health concern. While known to be strongly associated with a number of cardiovascular and pulmonary diseases and cancers, smoking also leads to a variety of cutaneous manifestations. Objective This article reviews the effects of cigarette smoking on the skin and its appendages. Methods A literature review was based on a MEDLINE search (1966–2004) for English-language articles using the MeSH terms cutaneous, dermatology, tobacco, skin, and smoking. An additional search was subsequently undertaken for articles related to smoking and associated mucocutanous diseases, with the focus on pathogenesis and epidemiologic data. Articles presenting the highest level of evidence and latest reports were preferentially selected. Results Smoking is strongly associated with numerous dermatologic conditions including poor wound healing, wrinkling and premature skin aging, squamous cell carcinoma, psoriasis, hidradenitis suppurativa, hair loss, oral cancers, and other oral conditions. In addition, it has an impact on the skin lesions observed in diabetes, lupus, and AIDS. The evidence linking smoking and melanoma, eczema, and acne is inconclusive. Anecdotal data exist on the possible protective effects of smoking in oral/genital aphthosis of Behçet's disease, herpes labialis, pyoderma gangrenosum, acral melanoma, and Kaposi's sarcoma in AIDS patients. Conclusions An appreciation of the adverse cutaneous consequences of smoking is important. Dermatologists can play an integral role in promoting smoking cessation by providing expert opinion and educating the public on the deleterious effects of smoking on the skin.
Article
Purpose Psoriatic skin lesions are associated with chronic inflammation related to immune cell activity. Therefore, the aim of this study was to compare changes in the proteome of psoriatic keratinocytes and lymphocytes. Experimental design A proteomics approach (nanoflow LC‐MS/MS) was used to analyze the expression of proteins in keratinocytes and lymphocytes from psoriatic patients and healthy controls. Results As a result 2119 proteins for keratinocytes and 1235 proteins for lymphocytes were identified, quantified, and assigned for further analysis. Psoriatic keratinocytes had 68 downregulated and 7 upregulated proteins and psoriatic lymphocytes had 106 downregulated and 67 upregulated proteins compared to healthy individuals. The list of proteins that expression was downregulated included proteins involved in antioxidant homeostasis, transcription regulation, and ATP hydrolysis; proteins with an upregulated expression were involved in glycolytic processes and RNA translation. These changes in both cell types were accompanied by an increased level of 4‐HNE‐protein adducts; control cells were characterized by 4‐HNE‐Lysine adducts formed with structural and binding proteins, while in psoriatic cells 4‐HNE‐Lysine, 4‐HNE‐Histidine, and 4‐HNE‐Cysteine adducts with various molecular function proteins occurred at a significant level. Conclusion This study highlighting the changes in psoriatic keratinocytes and lymphocytes that can be directly involved in the development of psoriasis. In both cell types the most significant changes were associated with upregulation of phosphoglycerate mutase 1 and downregulation of thioredoxin reductase. This article is protected by copyright. All rights reserved
Article
Skin represents the most extended organ of human body, having as main function the protection of our body from outdoor stressors. Its protective ability is compromised when the skin is disrupted as a consequence of mechanical insults. For this purpose, cutaneous tissue is equipped with an efficient and fine mechanism involved in repairing the wounded area. Among the numerous players that take part in the wound healing process, SR-B1 has been recently shown to have a role in keratinocyte re-epithelialization. SR-B1 is a mediator of cholesterol uptake from HDLs, whereas it is implicated in other cellular processes such as vitamins absorption, vesicle trafficking or pathogen identification. The aim of this study was to investigate the mechanisms involved in SR-B1 role in skin wound closure. Our in vitro data demonstrated that SR-B1 influenced keratinocyte proliferation and migration through a downregulation of nuclear cyclin D1 levels and active MMP9 expression respectively possibly in an NF-kB-dependent mechanism. In addition, SR-B1 was also able to modulate keratinocyte morphology into a pro-migratory cytoskeleton rearrangement. The present in vitro study suggests a new role of SRB1 as a possible new key player in cutaneous wound healing mechanism.
Article
Exposure to air pollution is associated with increased respiratory morbidities and susceptibility to lung dysfunction. Ozone (O3) is commonly recognized as one of the most noxious air pollutant and has been associated with several lung pathologies. It has been demonstrated that decreased lung disorder severity and incidence are connected with the consumption of a diet rich in fruits and vegetables, suggesting that higher intake of dietary micronutrients and phytoactive compounds can be beneficial. However, dietary supplementation - i.e. vitamin E (α-tocopherol) or vitamin A - has not always been effective in improving pulmonary function. Recently, research on the role of nutritional antioxidants on human health has focused more on studying their uptake at the cellular level rather than their effective ability to scavenge reactvive oxygen species (ROS). The Scavenger Receptor B1 (SRB1) has been shown to play a prominent role in the uptake, delivery and regulation of vitamin E in the lung. Given the importance of SRB1 in maintaining lung tissue in a healthy condition, we hypothesize that its expression could be modulated by pollution exposure, which thus could indirectly affect the uptake and/or delivery of lipophilic substances, such as vitamin E. To characterize the molecular mechanism involved in the redox modulation of SRB1, its cellular levels were assessed in human alveolar epithelial cells after O3 exposure. The results demonstrated that O3 induced the loss of SRB1 protein levels. This decline seems to be driven by hydrogen peroxide (H2O2) as a consequence of an increased activation of cellular NADPH oxidase (NOX), as demonstrated by the use of NOX inhibitors or catalase that reversed this effect. Furthermore, O3 caused the formation of SRB1-aldheyde adducts (4-hydroxy-2-nonenal) and the consequent increase of its ubiquitination, a mechanism that could account for SRB1 protein loss.
Article
Atopic dermatitis is the commonest chronic inflammatory skin disorder, affecting up to 20% of children and 10% of adults in industrialised countries. This highly debilitating condition poses a considerable burden to both the individual and society at large. The pathophysiology of atopic dermatitis is complex, encompassing both genetic and environmental risk factors. Dysregulation of innate and adaptive immunity plays a key role; however, recent epidemiological, genetic and molecular research has focused interest on skin barrier dysfunction as a common precursor and pathological feature. Current understanding of the aetiology of atopic dermatitis highlights disruption of the epidermal barrier leading to increased permeability of the epidermis, pathological inflammation in the skin, and percutaneous sensitisation to allergens. Thus, most novel treatment strategies seek to target specific aspects of the skin barrier or cutaneous inflammation. Several studies have also shown promise in preventing atopic dermatitis, such as the early use of emollients in high‐risk infants. This may have broader implications in terms of halting the progression to atopic comorbidities including food allergy, hay fever, and asthma. This article is protected by copyright. All rights reserved.
Article
Ozone-therapy initially applied in medicine by an empirical approach, has now reached a new stage where most of the biological mechanisms of ozone action have been clarified, that refers to antimicrobial effects, immunoregulation, antioxidant defenses and epigenetic modification. Current ozone medical preparation in dermatology mainly classified as ozone hydrotherapy, ozonated oil externally used and ozone autohemotherapy (OAHT). Admittedly, ozone is widely used in various fields against gram-negative and gram-positive bacteria, viruses, and fungi. More recently, great progress has been obtained in wound healing which is a multiphase process that consists of three overlapping but distinct stages: inflammation, tissue proliferation and remodeling. While the exact mechanisms of ozone-therapy still remain unclear. Therefore, more evidence is required before ozone can be presented as a promising method for the management and prevention of various skin diseases. In this review, we review the application status of ozone in dermatology and summarize possible mechanisms of ozone-therapy on skin diseases, aims to shed a light on providing a series of theoretical basis for its applications.