You are on page 1of 18

International Journal of Cosmetic Science, 2005, 27, 17–34

Ultraviolet radiation and skin aging: roles of reactive


oxygen species, inflammation and protease
activation, and strategies for prevention of
inflammation-induced matrix degradation – a review

S. Pillai, C. Oresajo and J. Hayward


Engelhard Corporation, Long Island, New York, NY, U.S.A.

Received 3 June 2004, Accepted 28 September 2004

Keywords: antipotease, elastase, matrix metalloprotease, neutrophils, serine protease

vation of matrix metalloproteases. The inflamma-


Synopsis
tion further activates the transcription of various
Inflammation and the resulting accumulation of matrixes degrading metalloproteases, leading to
reactive oxygen species (ROS) play an important abnormal matrix degradation and accumulation of
role in the intrinsic and photoaging of human skin non-functional matrix components. In addition,
in vivo. Environmental insults such as ultraviolet the inflammation and ROS cause oxidative damage
(UV) rays from sun, cigarette smoke exposure and to cellular proteins, lipids and carbohydrates,
pollutants, and the natural process of aging con- which accumulates in the dermal and epidermal
tribute to the generation of free radicals and ROS compartments, contributing to the aetiology of
that stimulate the inflammatory process in the photoaging. Strategies to prevent photodamage
skin. UV irradiation initiates and activates a com- caused by this cascade of reactions initiated by UV
plex cascade of biochemical reactions in human include: prevention of UV penetration into skin by
skin. In short, UV causes depletion of cellular anti- physical and chemical sunscreens, prevention/
oxidants and antioxidant enzymes (SOD, catalase), reduction of inflammation using anti-inflammatory
initiates DNA damage leading to the formation of compounds (e.g. cyclooxygenase inhibitors, inhibi-
thymidine dimmers, activates the neuroendocrine tors of cytokine generation); scavenging and
system leading to immunosuppression and release quenching of ROS by antioxidants; inhibition of
of neuroendocrine mediators, and causes increased neutrophil elastase activity to prevent extracellular
synthesis and release of pro-inflammatory media- matrix damage and activation of matrix metallo-
tors from a variety of skin cells. The pro-inflamma- proteases (MMPs), and inhibition of MMP expres-
tory mediators increase the permeability of sion (e.g. by retinoids) and activity (e.g. by natural
capillaries leading to infiltration and activation of and synthetic inhibitors).
neutrophils and other phagocytic cells into the
skin. The net result of all these effects is inflamma-
tion and free radical generation (both reactive oxy-
gen and nitrogen species). Furthermore, elastsases Résumé
and other proteases (cathepsin G) released from
neutrophils cause further inflammation, and acti- L’inflammation et l’accumulation de dérivés réac-
tifs de l’oxygène qui en résulte jouent un rôle
important sur le vieillissement normal (chronologi-
Correspondence: Dr Sreekumar Pillai PhD, Engelhard Cor-
poration, 50 Health Sciences Drive, Stony Brook, NY
que) et pathologique (photovieillissement) de la
11790, U.S.A. Tel.: +1 631 380 2352; fax: +1 631 380 peau in vivo. Les attaques de l’environnement, tels
2515; e-mail: kumar.pillai@engelhard.com que les UV, l’exposition a la fumée de cigarettes et

ª 2005 Society of Cosmetic Scientists and the Société Française de Cosmétologie 17


Ultraviolet radiation and skin aging S. Pillai, C. Oresajo and J. Hayward

a des polluants, et le procédé naturel du vieillisse- role in the intrinsic and photoaging of human skin
ment, contribuent à la création de radicaux-libres in vivo. Environmental insults such as ultraviolet
et de dérivés réactifs de l’oxygène qui stimulent (UV) rays from sun, cigarette smoke exposure and
l’inflammation cutanée. Les radiations UV sont à pollutants, as well as the natural process of aging
l’origine d’une cascade complexe de réactions bio- contribute to the generation of ROS that stimulate
chimiques sur la peau humaine. A court terme, les the inflammatory process in the skin. One of the
UV dégradent les antioxydants cellulaires et les primary events in ROS-mediated inflammation is
enzymes antioxydants (SOD, catalase); endomma- the activation of transcription factors that regulate
gent l’ADN provoquant la formation de dimères de the proteolytic degradation of the skin extracellu-
la thymidine; activent le système neuroendocrinien lar matrix (ECM). Photoaged skin shows increased
engendrant une immunodépression et la sécrétion proteolytic activation and abnormal ECM turnover
de médiateurs neuroendocriniens; et enfin provo- leading to increased degradation of collagen and
quent une synthèse accrue et la sécrétion de elastic fibres in the dermis, resulting in a loss of
médiateurs pro-inflammatoires par diverses cellules skin’s ability to resist stretching.
cutanées. Les médiateurs pro-inflammatoires aug- An interlinked network of enzymes that convert
mentent la perméabilité des capillaires engendrant ROS to harmless water and molecular oxygen
l’activation et l’infiltration de neutrophiles et regulates skin’s antioxidant defence system. The
autres cellules phagocytes dans la peau. Le résultat skin of aged and photoaged individuals has
de toutes ces réactions est l’inflammation et la reduced levels of these natural enzymatic and non-
génération de radicaux-libres (à la fois oxygénés et enzymatic antioxidant defence mechanisms,
azotés). Puis, les élastases et autres protéases increased neutrophil infiltration into skin and
(cathepsine G) issues des neutrophiles accroissent increased inflammation. The anti-aging technology
l’inflammation et activent les métalloprotéases used today in skin care products relies heavily on
matricielles (MPM). En effet, l’inflammation ainsi the use of antioxidants for prevention of UV- and
créée active la transcription de diverses MPM ROS-mediated inflammation. New basic under-
néfastes à la matrice extracellulaire, générant sa standing on the mechanisms of neutrophil activa-
dégradation anormale et l’accumulation de dérivés tion and emerging technologies on inhibition of
non fonctionnels en son milieu. De plus, l’inflam- UV- and ROS-induced proteases have significantly
mation et les dérivés réactifs de l’oxygène oxydent enhanced our ability to prevent UV-induced skin
et dégradent les protéines, lipides et sucres cellulai- aging.
res qui s’accumulent dans le derme et l’épiderme, Excellent reviews have been written on the free
contribuant à l‘étiologie du photovieillissement. radical theory of aging and the role of oxidative
Les stratégies pour prévenir de la photo-dégrada- stress and mitochondrial involvement in aging
tion générée par cette cascade de réactions causées process [1–3]. The purpose of this review is to
par les UV sont les suivantes: prévention de la focus on the role of inflammation, especially that
pénétration des UV dans la peau par des écrans caused by UV irradiation including neutrophil acti-
solaires physiques ou chimiques; prévention et vation and proteolytic activation in skin aging and
réduction de l’inflammation en utilisant des agents methods to prevent skin aging by interfering with
anti-inflammatoires (tels les inhibiteurs de la cyclo- these mechanisms.
oxygenase et les inhibiteurs de la production de la
cytokine); en complexant et bloquant les dérivés
Effects of UV on ROS generation,
réactifs de l’oxygène; inhibition de l’activité du
inflammation, neutrophil activation and
neutrophile élastase pour prévenir la dégradation
matrix metalloprotease-induced ECM
de la matrice extracellulaire et l’activation des
degradation
MPM; et inhibition de l’expression des MPM (ex:
par des rétinoı̈des) et de leur activité (ex: par des
UV, photoaging and skin changes
inhibiteurs naturels et synthétiques).
Skin changes with photoaging
Premature aging of the skin (photoaging) is a
Introduction
well-documented consequence of exposure to
Inflammation and the resulting accumulation of Ultraviolet A (UVA) and Ultraviolet B (UVB). Pho-
reactive oxygen species (ROS) play an important toaged skin is biochemically characterized by the

18 ª 2005 International Journal of Cosmetic Science, 27, 17–34


Ultraviolet radiation and skin aging S. Pillai, C. Oresajo and J. Hayward

presence of abnormal elastic fibres in the dermis cellular infiltration with neutrophils and mono-
and by a dramatic decrease of distinct collagen cytes within dermis and epidermis. Biochemical
types. The elastic fibres demonstrate a dystrophic, changes include release of histamine and accumu-
amorphic structure as a consequence of UV lation of cyclooxygenase- and lipooxygenase-
directly increasing tropoelastin gene expression derived products of arachidonic acid, kinins and
[4]. Photoaged skin is coarsely wrinkled and asso- cytokines derived from keratinocytes and fibro-
ciated with skin thickening (both epidermal and blasts. These responses are more pronounced in
dermal compartments), dyspigmentation and young subjects [18]. Aged skin demonstrated
telangiectasia [5–7]. Biochemically, the photoaged decreased epidermal turnover, diminished inflam-
skin shows increased proteolytic activation, and matory response to UV and impaired immune
abnormal ECM turnover leading to increased deg- function. In addition to the differences in immune
radation of collagen and elastic fibres in the der- and inflammatory responses of young versus aged
mis, resulting in a loss of skin’s ability to resist skin, the response of barrier function and barrier
stretching. Environmental insults such as UV rays, recovery is also impaired in older skin. In studies
cigarette smoke exposure and pollutants, and the with murine epidermis, it was demonstrated that
natural process of aging contribute to the genera- the skin becomes less sensitive to UVB-induced
tion of ROS that stimulate the inflammatory pro- barrier alterations with age and this decreased
cess in the skin. The skin of aged and photoaged sensitivity correlated with decreased DNA synthe-
individuals has reduced levels of natural enzymatic sis following UV irradiation [19].
and non-enzymatic antioxidant defence mecha- Acute and chronic UV exposure is associated
nisms [8] increased neutrophil infiltration into skin with protein oxidation in human skin in vivo, both
and increased inflammation [9]. in the living layers as well as in the stratum cor-
neum [20, 21]. Human dermal fibroblasts play a
Cellular mechanisms activated by UV irradiation central role in connective tissue breakdown in
The acute effects of UV irradiation on human skin photoaging. UV irradiation induces UV-responsive
are well characterized. The immediate reaction to genes of fibroblasts, among them matrix metallo-
UV is mediated by several mechanisms including proteases, which degrade macromolecules of the
direct effects on keratinocytes to release pro- ECM [22]. UVB modulates several signal transduc-
inflammatory cytokines such as interleukin-1 tion pathway(s) leading to generation of ROS that
(IL-1) and tumour necrosis factor-a (TNF-a) [10]; induces the transcription of metalloproteases
direct effects of photons on DNA to cause DNA [13, 23]. Recent data also suggest the potential
breakage [11]; depletion of cellular antioxidants involvement of reactive nitrogen species (RNS) in
and generation of ROS including hydrogen perox- addition to ROS in UVB-mediated expression of
ide, hydroxyl radical, singlet oxygen and peroxyl interstitial collagenase [matrix metalloprotease-1
radicals [5, 8]; and generation of prostaglandins (MMP-1)] and stromelysin-1 (MMP-3) [23]. One of
and other inflammatory mediators such as hista- the early responses of keratinocytes to UV irradi-
mine and leucotrines by mast cells in skin [12]. ation is release of pro-inflammatory cytokines in a
UV-induced inflammation and the resulting accu- similar fashion to response to injury.
mulation of ROS play an important role in the
intrinsic and photoaging of human skin in vivo
Release of inflammatory mediators in response to
[13]. At the cellular level, UV radiation triggers
UV
cytokine production [14], induces surface expres-
sion of adhesion molecules [15] and affects cellular Cytokines released in response to UV
mitosis, apoptosis and necrosis [16]. These media- UV, inflammation, tape stripping and even
tors also increase the skin’s sensitivity to irritants. mechanical stimulation of skin can cause release
The free radical-induced peroxidation of membrane of IL-1 from stratum corneum to initiate the
lipids contributes to increased phospholipase A2 inflammatory reactions within the living layers of
activity leading to more production of prostaglan- skin [10, 24]. In addition, UV irradiation stimu-
dins [17]. Morphologically acute UV irradiation is lated TNF-a synthesis and release from keratino-
followed by epidermal keratinocytes damage, cytes into blood stream, suggesting epidermal cell
depletion of Langerhans cells, dermal edema, derived cytokines may mediate systemic inflamma-
endothelial swelling, mast cell degranulation and tory reactions [25]. These two ‘primary cytokines’,

ª 2005 International Journal of Cosmetic Science, 27, 17–34 19


Ultraviolet radiation and skin aging S. Pillai, C. Oresajo and J. Hayward

can in turn induce the synthesis and release of reactions such as sunburn reaction and photo-
other pro-inflammatory cytokines in response to induced immune alterations [35]. Other cytokines
injury or UV radiation [14, 26, 27]. UV irradiation such as b-FGF, NGF, endothelin-1 and POMC-
dramatically induced the production and secretion derived peptides also stimulate melanocyte growth,
of IL-1, IL-3 and IL-6 that are normally low in un- melanogenesis and skin tanning [36]. Cytokines
irradiated cells. Additionally, UV irradiation of generated by the keratinocytes in the acne lesions
human skin in vivo also induced the expression of and comedonal part of acne may also influence
IL-1, IL-10 and IL-7 [28]. IL-10 and IL-12 are sebum production and inflammation of acne
regulated differentially by UVB irradiation [29]. lesions [37]. UV irradiation induced the production
UVB induced both IL-10 and IL-12, however, UVA of IL-1a, IL-1 receptor antagonist (IL-1 ra), Il-6
appears to induce only IL-10, not IL-12 in kera- and IL-10 in acne patients. UV-induced IL-1 ra
tinocytes. UV irradiation also induced the IL-10 and IL-10 may have a role as anti-inflammatory
and IL-12 expression in lymph nodes [30] suggest- regulators in acne. The levels of these two cyto-
ing the systemic role of UV in inflammatory kines were elevated in stationary acne patients,
responses. In addition to ILs, TNF-a and granulo- suggesting that UV irradiation may benefit the sta-
cyte–macrophage colony-stimulating factor (GM– tionary acne patients.
CSF) levels are also increased in response to UV
[31]. All these factors, in addition to mediating
ROS, inflammation and protease activation
inflammation, also cause immunosuppression.
Other growth factor agonists/antagonists secreted ROS generation and inflammation
by keratinocytes in response to UV include: IL-1 Generation of ROS is associated with life under
receptor antagonist [26]; alpha-melanocyte-stimu- aerobic conditions. ROS are generated under phy-
lating hormone (a-MSH) [32] vascular endothelial siological and pathological conditions. Superoxide,
growth factor (VEGF) [33]; nitric oxide (NO) [34, hydrogen peroxide and hydroxy radicals are pri-
35]; basic fibroblast growth factor (b-FGF); nerve marily produced by phagocytic cells and polynu-
growth factor (NGF), endothelin-1 and the proopi- clear lymphocytes in response to inflammation
omelanocortin (POMC)-derived peptides – MSH, [38]. Phagocytic cells such as macrophages and
ACTF, beta-LPH and beta-endorphin [36]. lymphocytes produce large quantities of ROS by
expressing high amounts of NADPH oxidase
Influence on other cell types in skin enzyme [39]. Involvement of NADPH oxidase-
Leukocyte/keratinocyte cell adhesion is mediated induced ROS in host defence response of these cells
by intercellular adhesion molecule-1 (ICAM-1) on is well established. Skin is uniquely vulnerable to
the keratinocyte cell surface. ICAM-1 serves as the the damage caused by ROS, being rich in unsatur-
counter-receptor for lymphocyte function associ- ated fatty acids and exposed to high oxygen ten-
ated antigen-1 for adhesion to keratinocytes [15]. sion and UV light, both of which promote ROS
IL-1a released in response to UV irradiation stimu- generation. Additionally, the respiratory burst of
lates the ICAM-1 expression of keratinocytes, infiltrating polymorphonuclear leukocytes and
thereby promoting the leukocyte adhesion to skin macrophages in inflamed skin produces high local
and generation or ROS by the leukocytes. a-MSH levels of superoxide and can release catalytic ion
produced by keratinocytes can in turn act as an from ferritin. The iron in combination with ROS
immunosuppresent or as a stimulant of melano- can cause additional damage to skin [38]. ROS,
cytes to produce more melanin for protection of especially the hydroxyl radical, are capable of
skin from further UV damage [32]. The immuno- damaging biological macromolecules such as
suppressive capacity of a-MSH is mediated by its DNA, carbohydrates, lipids and proteins. Among
effect on melanocyte and macrophage functions. the most susceptible targets are polyunsaturated
Another mediator of immunity and skin pigmenta- fatty acids, where ROS initiates the process of lipid
tion, nitric oxide is also produced in keratinocytes peroxidation. Damage to the lipids in the cellular
in response to UV exposure. UV stimulates the membrane can cause cellular damage, leakage of
expression of inducible nitric oxide synthase cellular components and ultimately cell death.
(iNOS), the enzyme that produces NO in response Lipid aldehydes produced during peroxidation can
to UV or inflammation. The NO produced by kera- also react with amino acid side chains and nucleic
tinocytes appears to play a role in UV-induced skin acids to cause further damage.

20 ª 2005 International Journal of Cosmetic Science, 27, 17–34


Ultraviolet radiation and skin aging S. Pillai, C. Oresajo and J. Hayward

ROS generated at the injury site play an import- Chronic Protease-Antiprotease


Inflammation Balance
ant role in modulating the inflammatory response
under acute and chronic injury conditions. One of Degrades
Anti-
the primary events in ROS-mediated inflammation Neutrophils Elastase proteases
+
is the activation of transcription factors. Nuclear IL-8
Inhibits

factor kappa B (NFjB) and activator protein-1


Inactivates TIMP
(AP-1) are transcription factors that are regulated Activation of macrophages,
keratinocytes
by cellular redox levels, and involved in regulation
of gene expression [40]. These factors are co-ordi- Activates
Degradation
Release of IL-1, TNF-a,
nately responsible for a huge range of extracellular prostaglandins and MMPs of ECM
leukotrines
signalling molecules responsible for inflammation,
tissue remodelling, oncogenesis and apoptosis, pro- Figure 1 Role of neutrophil elastase in inflammation
cesses that orchestrate many of the degenerative and matrix metalloprotease activation.
processes associated with aging [41]. UV exposure
also induces the expression of inducible cyclooxyge-
nase (COX-2) and lipooxygenase (LOX) in human Inflammation and neutrophil protease activation
skin, leading to increased production of pro-inflam- Damage to connective tissues is a major complica-
matory mediators such as prostaglandins and tion of the inflammatory response. Inflammatory
thromboxanes [35]. tissue injury contributes to the pathological chan-
ges in various tissues and organs including skin in
ROS and activation of neutrophils photodamage. [49]. Proteases released by leuko-
Phagocytic cells such as neutrophils and monocytes cytes play a major role in tissue injury associated
infiltrate into skin from capillaries in response to with inflammation (Fig. 1). Polymorphonuclear
UV-induced production of cytokines such as IL-1 or leukocyte (PMN) secretes lysosomal enzymes from
TNF-a [42]. Large amounts of IL-1 are stored in open phagocytic vacuoles regulated by cytoplasmic
stratum corneum, the top layer of human epidermis microtubules. Some of the major enzymes secreted
[43]. In addition to keratinocytes, the phagocytic by PMN in response to inflammation are neutro-
cells themselves secrete cytokines (TNF-a, IL-1, phil elastase, interstitial collagenase (MMP-1), and
IL-8, etc.) that further enhance recruitment of cathepsin G [46]. The serine protease, neutrophil
inflammatory cells [44]. The phagocytic cells recrui- elastase is activated in response to inflammatory
ted and activated by these inflammatory cytokines tissue injury. Serine proteases have substrate spe-
generate ROS and NO as part of their defence mech- cificities and substrate binding characteristics that
anisms. Both compounds can combine to form per- differ entirely from those of the MMPs, as discussed
oxynitrite, a reactive species capable of inducing in detail in Direct inhibition of MMPs section [49].
lipid peroxidation and damage [45]. In addition,
these cells generate a variety of other molecular Polymorphonuclear serine proteases
oxygen species (O2), ÆOH and H2O2) that assist in the Serine proteases have been classified by their sub-
killing of microorganisms. They also release oxida- strate specificity into three types: trypsin-like,
tion products of membrane fatty acids (e.g. arachi- chymotrypsin-like and elastase-like. Serine prote-
donate), which are converted to thromboxanes and ase elastase prefers substrates with small aliphatic
prostaglandin that are additional mediators of chains. The mode of action of serine proteases
inflammation [46]. Phagocytic cells induce the ker- involves the amino acid serine, which has a
atinocytes to synthesize and secrete elafin, an inhib- hydroxyl group that acts as a nucleophile for
itor of human neutrophil elastase, which eventually hydrolytic cleavage [49]. In contrast, a metal ion
limits the damage caused by the inflammatory neu- (usually zinc), coordinates and activates the target
trophils to skin [44]. Elafin (also known as skin- protein amide carbonyl for hydrolysis by MMPs.
derived anti-leukoprotease/trappin-2) is highly Therefore, serine protease elastase differs funda-
induced in inflamed skin such as in psoriasis, after mentally from MMP elastase. Several matrix com-
wounding and in skin tumours [47]. In addition to ponents such as collagens, gelatins, elastins,
being an inhibitor of elastase, elafin is also incorpor- fibronectin, laminin and glycosaminoglycans are
ated in the cornified envelope in psoriatic skin excellent substrates for both serine proteases and
lesions [48]. metalloproteases.

ª 2005 International Journal of Cosmetic Science, 27, 17–34 21


Ultraviolet radiation and skin aging S. Pillai, C. Oresajo and J. Hayward

Neutrophil (serine) elastase, which is released also suppress the expression of MMP [59]. The
during neutrophil infiltration of the epidermis, has complex interaction between proteases and their
been shown to induce hyperproliferation in inhibitors can regulate the ECM breakdown in neu-
keratinocytes in vitro and in vivo [50]. Neutrophil trophil-mediated inflammatory skin diseases.
infiltration has been demonstrated in hyperproli-
ferative skin conditions such as psoriasis, especially
Role of UV in MMP activation
in pustular psoriasis [51, 52]. The secreted ela-
stase may be one mechanism by which hyperpro- MMPs and their endogenous inhibitors, TIMPs
liferative skin conditions persist in psoriasis [51]. MMPs are a group of zinc-dependent endopeptidas-
Hyperproliferation of keratinocytes can be induced es capable of degrading ECM components and are
by neutrophil elastase both in vitro and in vivo, involved in the turnover and remodelling of the
and topical application of elastase inhibitors can dermis [60]. In normal skin, the MMPs are
prevent elastase-induced keratinocyte hyperprolif- expressed in very low levels and are kept in their
eration [50]. The mechanism by which elastase inactive form bound to endogenous inhibitors.
induces keratinocyte proliferation is believed to be Inflammation, UV irradiation and normal process
mediated by TGF-a stimulation and EGF receptor of aging can activate MMPs leading to increased
activation [53]. Inhibition of UV-induced neutro- matrix degradation.
phil elastase prevented UV-mediated skin tumour The MMP family of genes consists of at least
formation in hairless mice [54]. Another elastase two-dozen individual enzymes in human tissues.
activity, fibroblast elastase, also has been shown to Each is structurally distinct and has the ability to
be involved in skin wrinkle formation. Inhibition degrade a particular subset of matrix proteins
of this enzyme has been demonstrated to prevent [60]. MMP expression is organ-dependent, and dif-
UV-induced wrinkle formation in skin [55]. ferent MMPs are expressed in different develop-
mental stages and in different diseases. For
Protease–antiprotease balance example, MMPs are involved in tumour progres-
Metalloproteinases and serine proteases can work sion and inflammatory disorders. MMPs can be
in combination to directly attack most of the ele- broadly classified into four major classes depending
ments of the ECM including interstitial stroma and on substrate specificity and structural and expres-
basement membranes. In addition, these enzymes sion characteristics: collagenases, which break
activate one another through cleavage of the pro- down collagens (MMP-1 and MMP-8); gelatinases,
enzyme forms and can also cause degradation of (which degrade denatured collagens – MMP2,
protease inhibitors that maintains the appropriate MMP 9); stromelysins, which have broad spectrum
protease–antiprotease balance in normal skin specificity, and membrane bound MMPs, which are
(Fig. 1). For example, cathepsin G (a serine prote- located mainly on tumour cells. In addition to col-
ase) can activate MMP-8; human leukocyte ela- lagens, MMPs also degrade other ECM components
stase (a serine protease) can inactivate the major gelatine, laminin, fibronectin and elastin. Most
endogenous tissue inhibitors of matrix metallopro- skin cell types (fibroblasts, keratinocytes, melano-
teases (TIMPs); and MMP-8 and MMP-9 can inacti- cytes) synthesize MMPs. The activities of MMPs
vate a-1-protease inhibitor, the major endogenous within the skin are regulated by endogenous
inhibitor of human leukocyte elastase [56, 57]. inhibitors, small molecular weight proteins called
Thus, by contributing to protease activation and tissue inhibitor of metalloproteases (TIMPs). TIMPs
inactivating endogenous inhibitors, the two classes bind MMPs in a 1 : 1 ratio. Skin cells produce sev-
of proteases can skew the protease–antiprotease eral classes of TIMPs, and the activity of the MMPs
balance toward pathological tissue degradation. is determined by the balance of MMPs to TIMPs
While the protease–antiprotease balance is strictly within the cells. In addition, the activity and the
regulated under normal conditions, a breakdown of production of MMPs are regulated by various
the control mechanism occurs in various disease cytokines that are either generated within skin or
conditions characterized by excess serine protease enter skin from the blood. The collagen breakdown
activity. However, other elastase inhibitors, secre- that occurs during inflammation, wound healing,
tory leukocyte protease inhibitor and elafin are tumour invasion and chronological and photoag-
resistant to degradation by MMP-8 [58]. In addi- ing is regulated via cytokine modulation of MMP
tion, the secretory leukocyte protease inhibitor can expression. Prevention or blocking these conditions

22 ª 2005 International Journal of Cosmetic Science, 27, 17–34


Ultraviolet radiation and skin aging S. Pillai, C. Oresajo and J. Hayward

using anti-inflammatory drugs, antioxidants, sun- five-fold, however TIMP expression is either not
screens, etc. is an indirect way to prevent MMP altered or only slightly elevated by UV [66, 67].
activation and reduce matrix breakdown. Figure 2 below summarizes the roles of UV and
ROS and the possible mechanisms of regulation of
MMPs in skin MMP in skin.
Human skin expresses three distinct collagenases, MMP activity is regulated at multiple steps from
MMP-1, -8 and -13. Using UV-exposed organ cul- gene transcription to enzyme activation. The fol-
tures of human skin, it was demonstrated that lowing sections detail some of the critical steps in
although several MMPs including MMP-1 and -13 the regulation of MMPs.
are stimulated by UVB, only MMP-1 inhibition
using specific antibodies prevented UV-enhanced Transcriptional activation
collagen breakdown in this model [61]. MMP-8 is The regulatory sites in the MMP genes (5¢ flanking
also stimulated by UVB in human skin in vivo region) contain an AP-1 regulatory element. Sev-
[62]. UVB, even at suberythemal doses, induced eral growth factors and cytokines stimulate the
expression of MMP-1, -3 and -9 in normal human expression of AP-1 transcription factors (Jun and
epidermis [63]. In addition, UVA has been shown Fos) that form dimers, bind to the AP-1 binding site
to induce the expression of MMP-1 in dermal fibro- of MMP genes and activate their gene expression
blasts in vivo and MMP-1, -2 and -3 in cultured [68]. In addition to the AP-1 signalling system, the
fibroblasts. TGF-b signalling system and the NFjB activated sys-
MMP-1 initiates cleavage of fibrillar collagen tem also play roles in MMP induction in the skin.
types I and III in the dermis, which is then further The mechanisms by which the cytokines and
degraded by MMP-2 and -9 [60]. In addition, sim- growth factors regulate Jun and Fos involve three
ultaneous expression of MMP-2, -3 and -9 could distinct classes of protein kinases: mitogen-activated
result in degradation of non-collagenous compo- protein kinases (MAPKs), extracellular stimulus-
nents of dermal ECM, including basement mem- regulated kinases (ERKs) stress-activated protein
brane glycoproteins and proteoglycans. Solar kinase/Jun N-terminal kinases (SAPK/JNKs). NFjB
irradiation may exacerbate chronological aging by is especially important in UV- and inflammation-
further increasing the elevated levels of MMPs in induced activation of MMPs because of the role of
aged skin. Furthermore, MMPs in aged skin may ROS in activating NFjB. Generation of ROS via
already be in a more active state because of the activation of NADPH oxidase also is necessary for
decreased levels of TIMPs [64]. stimulation of MMP expression through the MAPK-
signalling pathways.
Molecular mechanisms of MMP activation
UV and ROS can activate the protein kinase cas- Zymogen activation
cades to stimulate MMP gene expression [63, 65]. Most MMPs are synthesized and secreted as latent
ROS also directly stimulate MMP zymogen activa- precursors or zymogens. These have to be proteo-
tion by inducing the cleavage of the cysteine resi- lytically activated in the extracellular space. The
due association with the Zn active site. It is
suspected that in addition to the ROS, RNS also
are capable of enhancing MMP activities [23].
UV
Selected transcription factors such as NFjB, AP-
1 and AP-2 are activated by UV irradiation [40]. Kinase Cascades
(MAPK,ERK, SAPK/JNK)
These molecular mechanisms operate in an inter- NFkB activation
MMP gene expression
dependent manner. For example, NFjB activation
can in turn lead to the expression of a variety of C fos C jun ProMMPs
genes involved in immunological and inflamma- 5’ AP-1 3’
Zymogen activation
tory responses, including genes that encode cyto- Collagenases
Gelatinases
kines, matrix degrading metalloproteases, adhesion MMP activity stromelysin

molecules and regulators of cell growth, differenti- Photoaging Imperfect repair Collagen breakdown
ation and cell death. MMPs are one of the
UV-responsive genes that degrade macromolecules Figure 2 Matrix metalloprotease regulation by UV irra-
of the ECM. UV induces MMP transcription by diation in skin.

ª 2005 International Journal of Cosmetic Science, 27, 17–34 23


Ultraviolet radiation and skin aging S. Pillai, C. Oresajo and J. Hayward

latent MMPs are kept inactive by a ‘cysteine released from sensory nerves innervating the skin
switch’ formed by the interaction of a conserved upon UV exposure. In addition, a variety of cells
cysteine residue in the propeptide with the Zn in in the skin produce increased neuroendocrine
the active site of the enzyme [69]. Disruption of hormones such as proopiomelanocortin (POMC)
the cys-Zn bond and the cleavage of the prodo- peptides and their receptors as well as neurotro-
main lead to a conformational change and activa- phins after UV exposure. Neuropeptides and neu-
tion of the pro-MMPs. Examples of proteolytic rohormones are capable of directly or indirectly
activation are the activation of collagenase mediating UV-induced cutaneous inflammation by
(MMP-1) by plasmin, trypsin, and stromelysin, and the induction of vasodilatation, plasma extravasa-
activation of collagenase 3 (MMP-13) by plasmin, tion, and augmentation of UV-induced cytokine,
stromelysin and gelatinase (MMP-2). The activa- chemokine, or cellular adhesion molecule expres-
tion cascade between MMPs and other proteases sion required for activation and trafficking of
form a complex network in regulating tissue pro- inflammatory cells into the inflamed tissue. Neu-
teolysis. ropeptides and neurotrophins may also play a
role in the repair of cutaneous UV injury.
Regulation of proteolytic activity of MMPs
The third step in regulation of MMP activity in
Protective role of UV
skin is their endogenous inhibition. In the natural
state, MMPs are bound to non-specific inhibitors The ability of UV to induce pigmentation in vivo
such as a-2 macroglobulin and a-1 antiprotease and in vitro is well documented, but the intracel-
or to specific inhibitors such as the TIMPs [70]. lular signals that trigger this response are poorly
Four different TIMPs have been identified. TIMP-1 understood. In addition to the roles of keratino-
and -2 are secreted in soluble form; TIMP-3 and cyte-produced NO and a-MSH (described in Influ-
-4 are bound to the ECM. TIMPs bind MMPs in a ence on other cell types in skin section) in
1 : 1 stoichiometry. There is a certain degree of protecting skin from further damage by UV, UV
specificity for TIMP towards different MMPs. For radiation also stimulates melanocytes directly to
example TIMP-1 potentially inhibits most MMPs produce more melanins. A variety of UV-induced
except MMP-2 and MMP-14. TIMP-2 inhibits most cytokines produced by keratinocytes such as
MMPs except MMP-9 [71]. The balance between basic fibroblast growth factor (b-FGF), nerve
TIMPs and MMPs is critical in determining the growth factor (NGF), endothelin-1 and the
degradative potential of cells in normal and patho- POMC-derived peptides MSH, ACTF, beta-LPH
logical conditions. This balance is altered in dis- and beta-endorphin stimulate melanocyte mitosis,
eased states such as cancer development, tumour increase melanogenesis, enhance melanocyte
invasion, angiogenesis and arthritis and in normal dendricity and prevent apoptotic cell death fol-
conditions such as foetal development and chrono- lowing UV injury [36]. A novel mechanism by
logical aging. which UV damage to cellular DNA induces skin
melanogenesis and skin tanning was elucidated
by pioneering work from Dr Gilchrest’s laborat-
Neuroendocrine system and UV-induced skin
ory [73]. At the molecular level, UV radiation
inflammatory responses
causes cellular DNA damage [36] and activates
UV irradiation also causes alterations in the skin cellular-signalling pathways [7]. Increasing DNA
immune system. There is increasing experimental repair after irradiation enhanced UV-induced
evidence that UV-induced skin inflammation is melanogenesis. This response comes from the
influenced by the sensory nervous system and small DNA fragment thymine dinucleotides
the neuroendocrine system in the skin [72]. The (pTpT), selectively excised during the repair of
resulting complex network of cytokines, chemo- UV-induced DNA photoproduct. pTpT stimulated
kines, neuropeptides, neuropeptide-degrading melanogenesis in mammalian pigment cells and
enzymes, neurohormones, and other inflamma- intact skin, mimicking the effects of UV irradi-
tory mediators are involved in the infiltration of ation on skin [11]. The mechanism by which
neutrophils into skin and cutaneous inflamma- pTpT activates melanogenesis appear to be medi-
tion. Neuropeptides such as substance P (SP) and ated by activation of p53 gene transcription,
calcitonin gene-related peptide (CGRP) are leading to nuclear accumulation of this protein

24 ª 2005 International Journal of Cosmetic Science, 27, 17–34


Ultraviolet radiation and skin aging S. Pillai, C. Oresajo and J. Hayward

and increasing the specific binding of this tran- effective in this region would be microfine titanium
scription factor to its DNA consensus sequence dioxide or zinc oxide of sufficiently small particle
[74]. Thymidine oligonucleotides also induced size (Table I).
protein kinase C beta levels suggesting that this Evidence that a full-UV spectrum sunscreen can
DNA excised fragment induces tanning by upreg- protect human skin against the biological changes
ulating the levels of p53 gene mediated at least occurring in photoaging was presented by work
in part by stimulating protein kinase cascade performed by L’Oreal [76]. Daily use of a cream
[75]. containing a photostable combination of UVB and
UVA absorbers providing a continuous absorption
through the entire UV spectrum protected skin of
Strategies to prevent photoageing and
volunteers from damage from solar simulated radi-
inflammation-induced matrix
ation (SSR) damage. Daily exposure to SSR for
degradation
6 weeks caused skin erythema, hyperpigmenta-
UV-induced inflammation and the resultant photo- tion, increased skin thickness because of elastolysis
aging can be prevented by various strategies out- and decrease in collagen, increase in MMP-2
lined below. expression and increased deposition of lysozyme
and a-1-antitrypsin on elastic fibres. Daily use of
the sunscreens prevented all these biological chan-
Prevention of UV penetration to skin
ges [76]. Prevention of UV penetration using other
Ultraviolet A radiation of wavelength 320– physical sunscreens should also prevent activation
400 nm penetrates the epidermis resulting in of MMPs in the epidermis and dermis by UVA and
damage to the dermis. This causes skin to age pre- UVB.
maturely, with effects that include roughening,
blotchiness, sagging and wrinkles. UVA also con-
Prevention of inflammation using antioxidants
tributes to the development of skin cancer but it is
or anti-inflammatories
only during the last decade that the damaging
effects of UVA have been fully appreciated. UVA Antioxidants
absorbers are chemicals that absorb radiation in Topical antioxidants can prevent generation of
the 320–360 nm region such as benzophenones, ROS and subsequent inflammatory reactions. ROS
anthranilates and dibenzoyl methanes. Microfine are generated either by the normal process of
zinc oxide is a physical blocker and is particularly metabolism whereby excess electrons generated in
effective in this region (Table I). the mitochondrial respiratory chain are donated to
Ultraviolet B is radiation between 290 and molecular oxygen to generate superoxide anions,
320 nm and causes sunburn and skin cancer. UVB or by UV-induced non-enzymatic conversion of
radiation inhibits DNA, RNA and protein synthesis molecular oxygen into superoxide anion radical.
and induces erythemal responses. The principal In addition, under certain circumstances UV and
UVB absorbers are para-aminobenzoic acid deriva- visible lights are capable of causing excitation of
tives, salicylates, cinnamates and camphor deriva- molecular oxygen in the skin to form highly react-
tives plus microfine titanium dioxide (Table I). ive singlet species.
Ultraviolet C radiation from 100 to 290 nm is An interlinked network of enzymes that convert
filtered out by the ozone layer and does not reach ROS to harmless water and molecular oxygen
the earth’s surface, hence the current concern regulates skin’s antioxidant defence system. How-
about ozone depletion. The only sunscreen that is ever, it is clear from recent studies that the levels

Table I Commonly used sunscreens in cosmetics and OTC products and their UV absorption range

UV spectrum (nm) Sunscreens approved for topical use

UVA (320–400) Benzophenones (benzophenone 3), anthranilates, dibenzoyl derivatives (avobenzone or parsol 1789)
UVB (290–320) PABA derivatives, salicylates (octyl salicylate, homosalate), cinnamates (octylmethxycinnamate)
UVC (100–290) Physical UV blockers such as microfine titanium dioxide, zinc oxide

ª 2005 International Journal of Cosmetic Science, 27, 17–34 25


Ultraviolet radiation and skin aging S. Pillai, C. Oresajo and J. Hayward

of these major antioxidant enzymes are decreased the effects of natural phenolic compounds and
with age and in photoaging [8]. In addition, the their role in the prevention of UV-induced skin
levels of endogenous antioxidants such as tocophe- damage [89]. A list of the natural polyphenolic
rols, ascorbic acid and glutathione, the activities of antioxidants is shown in the Table II. All these
enzymes that generate these antioxidants such as studies suggest that both dietary and topical anti-
glutathione reductase and glutathione peroxidase, oxidants could play a significant role in the reduc-
and the activities of other antioxidant enzymes tion of inflammatory responses.
that directly inactivate ROS such as superoxide
dismutase and catalase are all reduced in intrinsic Anti-inflammatories
aging and in photoaging in human skin [8, 20, Anti-inflammatory actives (both steroidal and non-
21, 77, 78]. Sun exposure variation because of steroidal) have been demonstrated to prevent skin
seasonal changes (summer versus winter) also inflammation caused by UV irradiation [90]. These
affects the levels of antioxidant enzyme activities agents generally act by suppressing the generation
in the stratum corneum. Levels of catalase in stra- of inflammatory agents such as prostaglandins,
tum corneum are lesser in summer compared with leukotrines or inflammatory cytokines. Among the
winter, although SOD did not follow this pattern most commonly used are topical retinoids, which
[79]. An acute exposure to UVA or UVB also have been shown to block the UV-induced activa-
reduced catalase activity without affecting the SOD tion of inflammation mediated by transcription fac-
activity [79]. The recovery of the catalase enzyme tors [91]. The recent introduction of several
after UV exposure occurred in 3–4 weeks, follow- analogues with increased potency and reduced
ing an age-dependent fashion. Recovery was faster irritation, retinoid therapy has become a common
in younger individuals than in older individuals treatment for reversing the signs of photoaging
[79]. In normal human skin, there is an antioxid- [92–94]. Other therapies include use of vitamin D
ant gradient in the stratum corneum with lower [95], corticosteroids and newly developed non-ste-
concentrations in the outer layers and higher con- roidal immunosuppressive and anti-inflammatory
centrations in the deeper layers. This gradient is agents such as tacrolimus and pimecrolimus
disrupted with UV and ozone exposure, suggesting [96, 97]. Although these agents are currently
that oxidative stress disrupts the antioxidant distri- administered mostly for inflammatory skin dis-
bution of skin [78]. eases, there is potential for their use as an anti-
As the levels of endogenous anti-oxidants such inflammatory in acute UV damage.
as reduced glutathione (GSH) decrease with age, it
was suggested that addition of exogenous antioxi-
Inhibition of gene activation of MMPs
dants might reduce inflammatory responses to UV
radiation and the aging process itself. Topical Among the most used therapies for prevention and
application of several biologically relevant antioxi- reversal of photodamage are topical retinoids
dants such as vitamin E, ascorbic acid and carote- [98–100]. Retinoids are a group of precursors of
noids have been shown to provide photoprotection retinoic acid that can be converted to retinoic acid
and reduce photodamage in human skin [80]. In within the skin under normal physiological condi-
addition, a variety of naturally occurring antioxid- tions. These include retinol, retinal, retinyl esters
ant compounds from plant origin have been dem- with various fatty acids and other glycosidic pre-
onstrated to be beneficial by quenching free cursors of retinoic acid Topical treatment with all-
radical generation and preventing photodamage in trans retinoic acid prevents UV induction of MMPs
skin [80, 81]. Using in vitro models it was demon- by multiple mechanisms. One mechanism is by
strated that the dietary antioxidant quercetin, prevention of UV-induced accumulation of c-Jun
reverses age-related GSH depletion and oxidative protein, which is required for MMP expression. In
stress [82]. In fact, several polyphenolic antioxi- addition all-trans retinoic acid may also stimulate
dants of plant origin have been shown to be bene- the breakdown of jun protein through ubiquitin–
ficial in preventing photodamage and skin proteasome-mediated pathways [101]. Topical
inflammation. These include polyphenols from all-trans retinoic acid prevented UVB induction of
green tea [9, 83, 84], grape seed [85], pomegran- MMP-1, -3 and -9. Retinoic acid has no effect on
ate [86] and a variety of other plant extracts erythema suggesting that the effect on gene expres-
[87, 88]. An excellent recent review summarizes sion of MMP is specific. Topical glucocorticoids

26 ª 2005 International Journal of Cosmetic Science, 27, 17–34


Table II Natural polyphenolic antioxidants and anti-inflammatory compounds

Compounds (and their class) Natural sources Biological activities, skin benefits
Ultraviolet radiation and skin aging

Caffeic acid ferulic acid Basil, thyme, taqragon, asafetida, Protects membranes from lipid peroxidation, protects skin from UV-induced erythema,
(cinnamic acid derivative) chia, apple, peanuts photoprotective agent
Quercetin (flavonoid) Evening primerose leaves, onion, tea A natural flavonoid, powerful antioxidant, MMP inhibitor, Protects skin antioxidant systems,
anti-inflammatory, protects UV-induced lipid peroxidation
Apigenin (flavonoid) Apples, beans, onions, broccoli, etc. Prevents UV-induced carcinogenesis, inhibits ODC activity, antioxidant
Genistein (isoflavone) Soybean, Lima bean Tumor protector, phytoestrogen, protects from UV-induced hydrogen peroxide generation,
reduce inflammation
Resveratrol (phytoalexin) Grape skin, grape seed, peanuts Protects from UV-induced tumor initiation, LOX, ODC and COX inhibitor,

ª 2005 International Journal of Cosmetic Science, 27, 17–34


protects from UV-induced lipid peroxidation, phytoestrogen,
Nordihydroguaiaretic acid Chaparral leaves Powerful antioxidant, MMP inhibitor, LOX and COX inhibitor
Carnosic acid, ursolic acid, Sage, rosemary, periwinkle, thyme Chemoprotective, elastase inhibitor
rosemarinic acid (triterpinoids)
Silymarin (flavonolignan) Milkthistle Inhibition of UV-induced erythema, COX inhibition, ODC induction,
prevents UV-induced infiltration of leukocytes, inhibits NFjB activation by UVB
Epicatechin, epicatechin-3-gallate, Tea leaves (Camellia sinesis) in Protects UV-induced carcinogenesis, UV protection, blocks tumor invasion,
epigallocatechin and epigallocatechin gallate general, prevents UV-induced edema, lipid peroxidation, general protection from UV-induced skin aging
green tea in particular
Procyanidins, proanthocyanidins, Red grape skin, eyebright Produced in plants to protect plant against UV radiation. Reduces hydrogen peroxide generation,
gallotannins and ellagotannins (tannins) decrease tumor incidence, inhibit induction of ODC, and epidermal DNA synthesis
Pycnogenol bark of pine tree (pinus maritime) Prevent UV-induced tumor induction, anti-inflammatory

COX, cyclooxygenase; LOX, lipooxygenase; MMP, matrix metalloprotease; NFjB, nuclear factor kappa B; ODC, ornithine decarboxylase; UV, ultraviolet.
S. Pillai, C. Oresajo and J. Hayward

27
Ultraviolet radiation and skin aging S. Pillai, C. Oresajo and J. Hayward

may also control MMP gene activation mediated plant terpenoids such as quercetin [109, 110];
by its anti-inflammatory actions [102]. As dis- polymethoxy flavonoid, nobiletin, from citrus
cussed in the UV, photoaging and skin changes depressa [111]; fucose and fucose-rich polysaccha-
section, prevention of UV penetration to skin using rides [112]; and some plant extracts such as spa-
sunscreens can also prevent MMP induction in tholobi caulis [91] euonymus alatus [113] and
response to UV [76]. Rhizoma notopterygii [114].
Several synthetic inhibitors of MMPs have been
synthesized. These include hydroxamic acid deriva-
Direct inhibition of MMPs
tives (Batimastat, Marimastat, AG 3340), butanoic
Many natural or synthetic inhibitors can directly acid analogues (BAY 12-9566) and others [115].
block MMPs (Table III). Natural products inhibit One of the most potent inhibitor so far described is
MMPs non-specifically. Synthetic compounds are Ilomastat (aka Galardin, GM-6001), a hydroxamic
designed for specific interaction with the enzymes, acid derivative of a pseudo-dipeptide. Ilomastat
mostly by chelating the Zn atom from the active inhibits MMPs in the nanomolar range by chelat-
site. Inhibition of MMPs using this strategy would ing the Zn metal ion from the enzyme active site
inhibit all the MMPs, without any specificity. This [116, 117]. Other drugs in development are Neov-
strategy carries a risk in that activities of some astat an active from shark cartilage (AE-941; from
MMPs are required for cleavage and removal of Aeterna Laboratories, Quebec, Canada) and a vari-
damaged ECM components. For example, collage- ety of synthetic inhibitors from British Biotech
nase-1 (MMP-1) is activated by UV and is respon- Pharmaceuticals Ltd, Oxford, UK [118].
sible for dermal matrix damage. However,
gelatinases (MMP-2 and -9) not only are not
Inhibitors of leucocyte proteases
induced by UV, their activity is needed to ‘clear
away’ the damage produced by MMP-1. Therefore, Considerable energy has been devoted to develop-
a more preferred strategy is to selectively inhibit ing inhibitors of serine proteinase elastase (human
certain MMPs (e.g. MMP-1) while sparing other leukocyte elastase or HLE) and Cathepsin G. Inhi-
MMPs (MMP-2 and -9). Currently such selective bition of neutrophil elastase could in addition to
inhibitors are not available; however, such a strat- blocking ECM degradation, also prevent the prote-
egy has been patented [103]. Recently, a biotech- ase–antiprotease imbalance observed in inflamma-
nologically derived human identical TIMP-2 has tion and photoaging.
been introduced as a cosmetic ingredient [104]. In vitro inhibition of human leukocyte elastase
Natural non-specific inhibitors include: flavo- by non-peptidic natural products of plant origin
noids [105, 106] avocado and soybean unsaponi- has been described [119, 120]. Pentacyclic triterp-
fyables [107]; green tea polyphenols [9, 108]; enoid metabolites of plant origin are inhibitors of

Table III Natural and synthetic matrix metalloprotease (MMP) inhibitors

Inhibitor (class) Mechanism/Specificity

Neovastat (shark cartilage fraction) General inhibitor of MMP, possibly by chelation of Zn


Galardin or Ilomastat Highly specific high affinity inhibitor of MMPs, chelates Zn from active site.
(hydroxamate derivative)
Batimastat, Marimastat, AG 3340 General MMP inhibitor, Zn chelation
(hydroxamate derivatives)
BAY 12-9566 (butanoic acid analogues Zn chelation
Green tea polyphenols Mode of action may be related to reducing inflammation; also directly
inhibits MMP by protein interaction
Several plant extracts Inhibition maybe by interfering with transcription regulation by interfering with reactive
oxygen species- or inflammation-mediated MMP gene induction
Quercetin Possibly by interfering with inflammation-induced MMP gene transcription activation
Nordihydroguaiaretic acid Same as above
Nobilin (citrus polyphenol) Same as above

28 ª 2005 International Journal of Cosmetic Science, 27, 17–34


Ultraviolet radiation and skin aging S. Pillai, C. Oresajo and J. Hayward

Table IV Leukocyte elastase inhibitors

Compound Sources Mechanism of inhibition

Ursolic acid and other plant terpenoid derivatives Apple, rosemary, basil, Holy basil Reversible inhibition
Fatty acids, bile acids Fish oils, vegetable oils Non-specific inhibition
Pyrene trisulfonic acid Synthetic Non-specific inhibition
N-Phenylphosphoonyl-leucyl-tryptophan Synthetic Specific inhibitor of skin fibroblast elastase

hydrolysis of both synthetic peptide substrates and tryptophane prevented this increase. These find-
elastin by HLE. Ursolic acid, the most potent of ings suggest that skin fibroblast elastase plays an
these compounds, has an inhibition constant of equally important role as HLE in mediating UV
4–6 lm for hydrolysis of peptide substrates [119]. effects on skin. Strategies to block both the HLE
Other non-specific inhibitors of HLE are fatty and fibroblast elastase would therefore provide
acids, bile acids, and pyrene trisulfonic acid enhanced protection.
(Table IV) [121–124]. These compounds are all
reversible inhibitors, and form no covalent interac-
Summary
tions with the enzyme. Because they bind at the
extended substrate-binding site, they are competit- UV irradiation initiates and activates a complex
ive inhibitors of enzymatic hydrolysis of larger cascade of biochemical reactions in human skin
oligopeptide substrates and proteins such as elas- (Fig. 3). In short, UV causes depletion of cellular
tin, but are non-competitive inhibitors of hydroly- antioxidants and antioxidant enzymes (SOD, cat-
sis of the smallest synthetic substrates, which bind alase), initiates DNA damage leading to the forma-
only in the immediate vicinity of the catalytic triad tion of thymidine dimmers, activates the
within the active site. neuroendocrine system leading to immunosuppres-
Specific inhibition of skin fibroblast elastase sion and release of neuroendocrine mediators and
activity has been demonstrated by a group at Kao causes increased synthesis and release of pro-
Corporation to block UV-induced wrinkle forma- inflammatory mediators from a variety of skin
tion [55]. Topical application of N-phenetylphos- cells. The pro-inflammatory mediators increase the
phonyl-leucyl-tryptophane, an inhibitor of skin permeability of capillaries leading to infiltration
fibroblast elastase appears to inhibit wrinkle for- and activation of neutrophils and other phagocytic
mation by UV in rats. Six weeks of UVB irradiation cells into the skin. The net result of all these effects
significantly enhanced elastase activity in rat skin, is inflammation and free radical generation (both
whereas topical N-phenetylphosphonyl-leucyl- reactive oxygen and nitrogen species).

Protective Neuroendocrine Release of


Inflammatory mediators ROS generation
role release

DNA damage
UV IL-1 alpha and TNF-a release
from keratinocytes
Activation of skin
Neuroendocrine system Synthesis and release of prostaglandins
Thymidine And release of neuropeptides, leucotrines, secondary cytokines
Dimer formation histamines (IL-6, IL-8) from skin cells Depletion of
cellular antioxidants

Melanocyte Inflammation Activation of neutrophils


Macrophages
activation
Cellular protein, lipid
Activation of transcription and CHO oxidation
Free Radical Generation
Factors (AP-1, AP-1, NFkB) Accumulation of ROS, NOS
Skin tanning/
darkening
MMP transcription Release of Neutrophil
activation elastase
Cell death,
Protection from lipid peroxidation
further UV damage Abnormal matrix Disruption of
Figure 3 A summary of the differ- degradation Protease-antiprotease
balance
ent mechanisms initiated by UV that
Accumulation of non-
Accumulation of
lead to reactive oxygen species gen- Functional matrix components
abnormal proteins,
cellular debris
eration, inflammation and photoag-
ing in skin. Photoaging

ª 2005 International Journal of Cosmetic Science, 27, 17–34 29


Ultraviolet radiation and skin aging S. Pillai, C. Oresajo and J. Hayward

Elastases and other proteases (cathepsin G) human skin in vivo. J. Invest. Dermatol. 117, 1212–
released from neutrophils cause further inflamma- 1217 (2001).
tion and activation of matrix metalloproteases. 9. Katiyar, S.K. and Mukhtar, H. Green tea polyphenol
The inflammation further activates the transcrip- (-)-epigallocatechin-3-gallate treatment to mouse
skin prevents UVB-induced infiltration of leukocytes,
tion of various matrix degrading metalloproteases,
depletion of antigen-presenting cells, and oxidative
leading to abnormal matrix degradation and accu-
stress. J. Leukoc. Biol. 69, 719–726 (2001).
mulation of non-functional matrix components. 10. Wood, L.C., Elias, P.M., Calhoun, C. et al. Barrier
Furthermore, the inflammation and ROS cause disruption stimulates interleukin-1 alpha expres-
oxidative damage to cellular proteins, lipids and sion and release from a pre-formed pool in mu-
carbohydrates, which accumulates in the dermal rine epidermis. J. Invest. Dermatol. 106, 397–403
and epidermal compartments, contributing to the (1996).
aetiology of photoaging. 11. Gilchrest, B.A. and Eller, M.S. DNA photodamage
Strategies to prevent photodamage caused by stimulates melanogenesis and other photoprotec-
this cascade of reactions initiated by UV include: tive responses. J. Investig. Dermatol. Symp. Proc. 4,
prevention of UV penetration into skin by physical 35–40 (1999).
12. Gonzalez, S. and Pathak, M.A. Inhibition of UV
and chemical sunscreens, prevention/reduction of
induced formation of ROS, lipid peroxidation, ery-
inflammation using anti-inflammatory compounds
thema and skin photosensitization by polypodium
(e.g. cyclooxygenase inhibitors, inhibitors of cyto- leucotomos. Photodermatol. Photoimmunol. Pho-
kine generation); scavenging and quenching of tomed. 12, 45–56 (1996).
ROS by antioxidants; inhibition of neutrophil ela- 13. Kawaguchi, Y., Tanaka, H., Okada, T. et al. The
stase activity to prevent ECM damage and activa- effects of UV A and reactive oxygen species on the
tion of MMPs, and inhibition of MMP expression mRNA expression of type IV collagenase and its tis-
(e.g. by retinoids) and activity (e.g. by natural and sue inhibitor in cultured human dermal fibroblasts.
synthetic inhibitors). Arch. Dermatol. Res. 288, 39–44 (1996).
14. Takashima, A. and Bergstresser, P.R. Impact of UV
radiation on the epidermal cytokine network. Phot-
References ochem. Photobiol. 63, 397–400 (1996).
15. Krutmann, J. and Grewe, M. Involvement of cytok-
1. Melov, S. Mitochondrial oxidative stress: physiolog-
ines, DNS damage and reactive oxygen intermedi-
ic consequences and potential for a role in aging.
ates in UV induced modulation of intercellular
Ann. NY Acad. Sci. 908 219–225 (2000).
adhesion molecule-1 expression. J. Invest. Dermatol.
2. Ashok, B.T. and Ali, R. The aging paradox: free
105 (Suppl. 1), 67S–70S (1995).
radical theory of aging. Exp. Gerontol. 34, 293–303
16. Bielenberg, D.R., Bucana, C.D., Sanchez, R. et al.
(1999).
Molecular regulation of UVB induced cutaneous
3. Wickens, A.P. Ageing and the free radical theory.
angiogenesis. J. Invest. Dermatol. 111, 864–872
Respir. Physiol. 128, 379–391 (2001).
(1998).
4. Bernstein, E.F., Brown, D.B., Urback, F. et al. UV
17. Hruza, L.L. and Pentland, A.P. Mechanisms of UV-
radiation activates the human elastin promoter in
induced inflammation. J. Invest. Dermatol. 100,
transgenic mice: a novel in vivo and in vitro model
35S–41S (1993).
of cutaneous photoaging. J. Investig. Dermatol. 105,
18. Soter, N.A. Acute effects of UV on the skin. Semin.
269–273 (1995).
Dermatol. 9, 11–15 (1990).
5. Lavker, R.M. Structural alterations in exposed and
19. Haratake, A., Uchida, Y., Mimura, K., Elias, P.M.
unexposed aged skin. J. Invest. Dermatol. 73, 559–
and Holleran, W.M. Intrinsically aged epidermis
566 (1979).
displays diminished UVB-induced alterations in bar-
6. Lavker, R.M. and Kligman, A.M. Chronic helio-
rier function associated with decreased prolifer-
dermatitis: a morphological evaluation of chronic
ation. J. Invest. Dermatol. 108, 319–323 (1997).
actinic dermal damage with emphasis on the role
20. Thiele, J.J. Oxidative targets in the stratum cor-
of mast cells. J. Invest. Dermatol. 90, 325–330
neum, a new basis for antioxidative strategies.
(1988).
Skin Pharmacol. Appl. Skin Physiol. 1, 87–91
7. Gilchrest, B.A. Skin aging and photoaging: an over-
(2001).
view. J. Am. Acad. Dermatol. 21, 610–613 (1989).
21. Sander, C.S., Chang, H., Salzmann, S. et al. Photo-
8. Rhie, G., Shin, M., Seo, J.Y. et al. Aging and photo-
aging is associated with protein oxidation in
aging dependent changes of enzymic and non-enzy-
human skin in vivo. J. Invest. Dermatol. 118, 618–
mic antioxidants in the epidermis and dermis of
625 (2002).

30 ª 2005 International Journal of Cosmetic Science, 27, 17–34


Ultraviolet radiation and skin aging S. Pillai, C. Oresajo and J. Hayward

22. Kang, S., Fisher, G.J. and Voorhees, J.J. Photoaging: 35. Seo, S.J., Choi, H.G., Chung, H.J. and Hong, C.K.
pathogenesis, prevention, and treatment. Clin. Geri- Time course of expression of mRNA of inducible
atr. Med. 17, 643–659 (2001). nitric oxide synthase and generation of nitric oxide
23. Brenneisen, P., Sies, H. and Scharffetter-Kochanek, by UV B in keratinocytes cell lines. Br. J. Dermatol.
K. UV-B irradiation and matrix metalloproteinases: 147, 655–662 (2002).
from induction via signaling to initial events. Ann. 36. Gilchrest, B.A., Park, H.Y., Eller, M.S. and Yarr,
NY Acad. Sci. 973, 31–43 (2002). M. Mechanisms of UV induced pigmentation. Photo-
24. Kupper, T.S. and Groves, R.W. The interleukin-1 chem. Photobiol. 63, 1–10 (1996).
axis and cutaneous inflammation. J. Investig. Der- 37. Suh, D.H., Kwon, T.E. and Youn, J.I. Changes of
matol. 105, 62S–66S (1995). comedonal cytokines and sebum secretion after UV
25. Kock, A., Schwarz, T., Kirnbauer, R. et al. Human irradiation in acne patients. Eur. J. Dermatol. 12,
keratinocytes are a source for tumor necrosis factor 139–144 (2002).
alpha: evidence for synthesis and release upon sti- 38. Trenum, C.W., Blake, D.R. and Morris, C.J. Skin
mulation with endotoxin or UV light. J. Exp. Med. inflammation: reactive oxygen species and the role
172, 1609–1614 (1990). of iron. J. Invest. Dermatol. 99, 675–682 (1992).
26. Hirao, T., Aoki, H., Yoshida, T., Sato, Y. and Ka- 39. Chlopicki, S., Olszanecki, R., Janiszewski, M. et al.
moda, H. Elevation of interleukin-1 receptor antag- Antioxid. Redox Signal. 6, 691–698 (2004).
onist in the stratum corneum of sun-exposed and 40. Abeyama, K., Eng, W., Jester, J.V. et al. A role for
UV B-irradiated human skin. J. Invest. Dermatol. NF-kappa B dependent gene transactivation in sun-
106, 1102–1107 (1996). burn. J. Clin. Invest. 105, 1751–1759 (2000).
27. Shreedhar, V., Giese, T., Sung, V.W. and Ullrich, 41. Rittie, L. and Fisher, G.J. UV-light-induced signal
S.E. A cytokine cascade including prostaglandin E2, cascades and skin aging. Ageing Res Rev. 1, 705–
IL-4, and IL-10 is responsible for UV-induced sys- 720 (2002).
temic immune suppression. J. Immunol. 160, 42. Krishnadasan, B., Naidu, B.V., Byrne, K. et al. The
3783–3789 (1998). role of proinflammatory cytokines in lung ischemia-
28. Brink, N., Szamel, M., Young, A.R., Wittern, K.P. reperfusion injury. J. Thorac. Cardiovasc. Surg. 125,
and Bergemann, J. Comparative quantification of 261–272 (2003).
IL-1 beta, IL-10, IL-10r, TNF alpha and IL-7 43. Hales, J.M. and Camp, R.D. Potent T cell stimulato-
mRNA levels in UV-irradiated human skin in vivo. ry material with antigenic properties in stratum
Inflamm. Res. 49, 290–296 (2000). corneum of normal human skin. J. Invest. Dermatol.
29. Kondo, S., The roles of keratinocyte-derived cytok- 110, 725–729 (1998).
ines in the epidermis and their possible responses to 44. Tanaka, N., Fujioka, A., Tajima, S., Ishibashi,
UVA-irradiation. J. Investg. Dermatol. Symp. Proc. 4, A. and Hirose, S. Elafin is induced in epidermis in
177–183 (1999). skin disorders with dermal neutrophilic infiltration:
30. Katiyar, S.K. Skin photoprotection by green tea: interleukin-1 beta and tumor necrosis factor-alpha
antioxidant and immunomodulatory effects. Curr. stimulate its secretion in vitro. Br. J. Dermatol. 143,
Drug Targets Immune Endocr. Metabol. Disord. 3, 728–732 (2000).
234–242 (2003). 45. Drew, B. and Leeuwenburgh, C. Aging and the role
31. Schwarz, T. and Luger, T.A. Effect of UV irradiation of reactive nitrogen species. Ann. NY Acad. Sci.
on epidermal cell cytokine production. J. Photochem. 959, 66–81 (2002).
Photobiol. B 4, 1–13 (1989). 46. Weissmann, G., Smolen, J.E. and Hoffstein, S. Poly-
32. Luger, T.A., Scholzen, T. and Grabbe, S. The role of morphonuclear leukocytes as secretory organs of
alpha melanocyte stimulating hormone in cutane- inflammation. J. Investig. Dermatol. 71, 95–99
ous biology. J. Investig. Dermatol. Symp. Proc. 2, (1978).
87–93 (1997). 47. Pol, A., Pfundt, R., Zeeuwen, P., Molhuizen, H. and
33. Mildner, M., Weninger, W., Trautinger, F., Ban, J. Schalkwijk, J. Transcriptional regulation of the ela-
and Tschachler, E. UVA and UVB radiation differ- fin gene in human keratinocytes. J. Invest. Derma-
entially regulate vascular endothelial growth factor tol. 120, 301–307 (2003).
expression in keratinocyte-derived cell lines and in 48. Nakane, H., Ishida-Yamamoto, A., Takashashi, H.
human keratinocytes. Photochem. Photobiol. 70, and Ilzuka, H. Elafin, a secretory protein, is cross-
674–679 (1999). linked into the cornified cell envelopes from the
34. Lee, S.C., Lee, J.W., Jung, J.E. et al. Protective role inside of psoriatic keratinocytes. J. Invest. Dermatol.
of nitric oxide-mediated inflammatory response 119, 50–55 (2002).
against lipid peroxidation in UVB-irradiated skin. 49. Simon, S.R., Roemer, E.J., Golub, L.M. and Rama-
Br. J. Dermatol. 142, 653–659 (2000). murthy, N.S. Serine proteinase inhibitory activity

ª 2005 International Journal of Cosmetic Science, 27, 17–34 31


Ultraviolet radiation and skin aging S. Pillai, C. Oresajo and J. Hayward

by hydrophobic tetracycline. US Patent no. 63. Fisher, G.J., Datta, S.C., Talwar, H.S. et al. Molecu-
5773430 (1998). lar basis of sun-induced premature skin ageing and
50. Rogalski, C., Ulf, M.-H., Proksch, E. and Wiedow, retinoic antagonism. Nature 379, 335–339 (1996).
O. Human leukocyte elastase induces keratinocyte 64. Ashcroft, G.S., Herrick, S.E., Tarnuzzer, R.W. et al.
proliferation in vitro and in vivo. J. Invest. Dermatol. Human ageing impairs injury-induced in vivo
118, 49–54 (2002). expression of tissue inhibitor of matrix metallopro-
51. Piskin, G., Tursen, U., Bos, J.D. and Teunissen, M.B. teinases (TIMP)-1 and -2 proteins and mRNA.
IL-4 expression by neutrophils in psoriasis lesional J. Pathol. 183, 169–176 (1997).
skin upon high-dose UVB exposure. Dermatology 65. Scharffetter, K., Wiaschek, M., Hogg, A. et al. UVA
207, 51–53 (2003). irradiation induces collagenase in human dermal fi-
52. Schaerli, P., Britschgi, M., Keller, M. et al. Charac- broblasts in vitro and in vivo. Arch. Dermatol. Res.
terization of human T cells that regulate neutrophi- 283, 506–511 (1991).
lic skin inflammation. J. Immunol. 173, 2151– 66. Herrmann, G., Wlasehek, M., Lange, T.S. et al. UVA
2158 (2004). irradiation stimulates the synthesis of various mat-
53. Meyer-Hoffert, U., Wingertszahn, J. and Wiedow, O. rix metalloproteinases in cultured human fibro-
Human leukocyte elastase induces keratinocyte pro- blasts. Exp. Dermatol. 2, 92–97 (1993).
liferation by epidermal growth factor receptor activa- 67. Petersen, M.J., Hansen, C. and Craig, S. Ultraviolet
tion. J. Investig. Dermatol. 123, 338–345 (2004). A irradiation stimulates collagenase production in
54. Starcher, B., O’Neal, P., Granstein, R.D. and Beis- cultured human fibroblasts. J. Invest. Dermatol. 99,
sert, S. Inhibition of neutrophil elastase suppresses 440–444 (1992).
the development of skin tumors in hairless mice. 68. Mauviel, A. Cytokine regulation of metalloprotein-
J. Invest. Dermatol. 107, 159–163 (1996). ase gene expression. J. Cell Biochem. 53, 288–295
55. Tsukahara, K., Takema, Y., Moriwaki, S. et al. (1993).
Selective inhibition of skin fibroblast elastase elicits 69. Van Wart, H.E. and Birkedal-Hansen, H. The cys-
a concentration-dependent prevention of UV B- teine switch: a principle of regulation of metallopro-
induced wrinkle formation. J. Invest. Dermatol. 117, teinase activity with potential applicability to the
671–677 (2001). entire matrix metalloproteinase gene family. Proc.
56. Mallya, S.A., Hall, J.E., Lee, H.M. et al. Interaction of Natl Acad. Sci. USA 87, 5578–5582 (1990).
matrix metalloproteinases with serine protease 70. Birkedal-Hansen, H. Moore, W.G.I., Bodden, M.K.
inhibitors. Ann. NY Acad. Sci. 732, 303–314 (1994). et al. Matrix metalloproteinases: a review. Crit. Rev.
57. Rinehart, A.R., Mallya, S., Simon, S.R. et al. Oral Biol. Med. 4, 197–250 (1993).
Human alpha-proteinase inhibitor binds to extracel- 71. Watson, S.A. and Tierney, G. Matrix metalloprote-
lular matrix in vitro. Am. J. Respir. Cell Mol. Biol. 9, inase inhibitors: a review. BioDrugs 9, 325–335
666–679 (1993). (1998).
58. Henry, M.T., McMahon, K., Costello, C., Fitz- 72. Scholzen, T.E., Brzoska, T., Kalden, D.H. et al.
gerald, M.X. and O’Connor, C.M. Secretory leuko- Effect of UV light on the release of neuropeptides
cyte proteinase inhibitor and elafin are resistant and neuroendocrine hormones in the skin: media-
to degradation by MMP-8. Exp. Lung Res. 28, tors of photodermatitis and cutaneous inflamma-
85–97 (2002). tion. J. Investig. Dermatol. Symp. Proc. 4, 55–60
59. Zhang, Y., DeWitt, D.L., McNeely, T.B., Wahl, S.M. (1999).
and Wahl, L.M. Secretory leukocyte protease inhib- 73. Eller, M.S., Ostrom, K. and Gilchrest, B.A. DNA
itor suppresses the production of monocyte PGH damage enhances melanogenesis. Proc. Natl Acad.
synthase-2, PGE2 and MMPases. J. Clin. Invest. 99, Sci. USA 93, 1087–1092 (1996).
894–900 (1997). 74. Eller, M.S., Maeda, T., Magnoni, C., Atwal, D. and
60. Kahari, V.M. and Saarialho-Kere, U. Matrix metal- Gilchrest, B.A. Enhancement of DNA repair in
loproteinases in skin. Exp. Dermatol. 6, 199–213 human skin cells by thymidine dinucleotides: evi-
(1997). dence for a p53-mediated mammalian SOS
61. Brennan, M., Bhatti, H., Nerusu, K.C. et al. Matrix response. Proc. Natl Acad. Sci. USA 94, 12627–
metalloproteinase-1 is the major collagenolytic 12632 (1997).
enzyme responsible for collagen damage in UV-irra- 75. Wu, C., Marks, S., Prk, H.Y., Howard, C. and Gilch-
diated human skin. Photochem. Photobiol. 78, rest, B.A. T-oligos induce mitf, tyrosinase and pkc-
43–48 (2003). beta in cultured human melanocytes. Pigment Cell
62. Fisher, G.J., Choi, H.C., Bata-Csorgo, Z. et al. UV Res. 17, 452 (2004).
irradiation increases matrix metalloproteinase-8 76. Seite, S., Colige, A., Piquemal-Vivenot, P. et al. A
protein in human skin in vivo. J. Invest. Dermatol. full-UV spectrum absorbing daily use cream pro-
117, 219–226 (2001). tects human skin against biological changes occur-

32 ª 2005 International Journal of Cosmetic Science, 27, 17–34


Ultraviolet radiation and skin aging S. Pillai, C. Oresajo and J. Hayward

ring in photoaging. Photodermatol. Photoimmunol. and DNA cleavage protectors. Phytomedicine 8,


Photomed. 16, 147–155 (2000). 125–132 (2001).
77. Thiele, J.J. Photoaging is associated with protein 89. Svobodova, A., Psotova, J. and Walterova, D. Nat-
oxidation in human skin in vivo. J. Invest. Dermatol. ural phenolics in the prevention of UV-induced skin
118, 618–625 (2002). damage: a review. Biomed. Pap. 147, 137–145
78. Weber, S.U., Thiele, J.J., Cross, C.E. and Packer, L. (2003).
Vitamin C, uric acid, and glutathione gradients in 90. Bissett, D.L., Chatterjee, R. and Hannon, D.P. Pho-
murine stratum corneum and their susceptibility to toprotective effect of topical anti-inflammatory
ozone exposure. J. Invest. Dermatol. 113, 1128– agents against UV radiation-induced chronic skin
1132 (1999). damage in the hairless mouse. Photodermatol.
79. Hellemans, L., Corstjens, H., Neven, A., Declercq, L. Photoimmunol. Photomed. 7, 153–158 (1990).
and Maes, D. Antioxidant enzyme sactivity in 91. Kang, I.C., Kim, S.A., Song, G.Y. et al. Effects of the
human stratum corneum shows seasonal variation ethyl acetate fraction of Spatholobi caulis on tumor
with an age-dependent recovery. J. Invest. Dermatol. cell aggression and migration. Phytother. Res. 17,
120, 434–439 (2003). 163–167 (2003).
80. Dreher, F. and Maibach, H. Protective effects of top- 92. Thacher, S.M., Vasudevan, J., Tsang, K.Y., Nagpal,
ical antioxidants in humans. Curr. Probl. Dermatol. S. and Chandraratna, R.A. New dermatological
29, 157–164 (2001). agents for the treatment of psoriasis. J. Med. Chem.
81. Podda, M. and Grundmann-Kollmann, M. Low 44, 281–297 (2001).
molecular weight antioxidants and their role in skin 93. Verschoore, M., Bouclier, M., Czernielewski, J. and
ageing. Clin. Exp. Dermatol. 26, 578–582 (2001). Hensby, C. Topical retinoids, their uses in dermatol-
82. Skaper, S.D., Fabris, M., Ferrari, V., Dalle Carbon- ogy. Dermatol. Clin. 11, 107–115 (1993).
are, M. and Leon, A. Quercetin protects cutaneous 94. Guenther, L.C. Topical tazarotene therapy for psori-
tissue-associated cell types including sensory neu- asis, acne and photoaging. Skin Therapy Lett. 7,
rons from oxidative stress induced by glutathione 1–4 (2002).
depletion: cooperative effects of ascorbic acid. Free 95. Griffiths, C.E. Retinoids and vitamin D analogues:
Radic. Biol. Med. 22, 669–678 (1997). action on nuclear transcription. Hosp. Med. 59,
83. Katiyar, S.K., Afaq, F., Perez, A. and Mukhtar, 12–16 (1998).
H. polyphenol (-)-epigallocatechin-3-gallate treat- 96. Gupta, A.K. and Chow, M. Pimecrolimus: a review. J.
ment of human skin inhibits UV radiation-induced Eur. Acad. Dermatol. Venereol. 17, 493–503 (2003).
oxidative stress. Carcinogenesis 22, 287–294 97. Niedner, R. Topical corticosteroids versus topical
(2001). inhibitors of calcineurin. Hautarzt 54, 338–341
84. Vayalil, P.K., Elmets, C.A. and Katiyar, S.K. Treat- (2001).
ment of green tea polyphenols in hydrophilic cream 98. Voorhees, J.J. and Fisher, G.J. Method of inhibiting
prevents UVB-induced oxidation of lipids and photoaging of skin. US Patent no. 5837224
proteins, depletion of antioxidant enzymes and (1998).
phosphorylation of MAPK proteins in SKH-1 hair- 99. Fisher, G.J., Voorhees, J.J. and Kang, S. Composi-
less mouse skin. Carcinogenesis 24, 927–936 tions and methods for inhibiting photoaging of
(2003). skin. US Patent no. 6365630 (2002).
85. Zhao, J., Wang, J., Chen, Y. and Agarwal, R. Anti- 100. Varani, J., Fisher, G.J., Voorhees, J.J. and Kang, S.
tumor-promoting activity of a polyphenolic fraction Methods and compositions for preventing and treat-
isolated from grape seeds in the mouse skin ing chronological aging in human skin. US Patent
two-stage initiation-promotion protocol and identifi- application no. 20010053347 (2001).
cation of procyanidin B5-3¢-gallate as the most 101. Fisher, G.J., Talwar, H.S., Lin, J. and Voorhees, J.J.
effective antioxidant constituent. Carcinogenesis 20, Molecular mechanisms of photoaging in human
1737–1745 (1999). skin in vivo and their prevention by all-trans reti-
86. Afaq, E., Saleem, M., Brans, R. and Mukhtar, H. noic acid. Photochem. Photobiol. 69, 154–157
Novel agents and targets for skin cancer chemopre- (1999).
vention: studies with pomegranate fruit extract. 102. Mauviel, A., Halcin, C., Vasiloudes, P., Prks, W.C.,
J. Investig. Dermatol. 121 (2003) (abstract no. Kurkinen, M. and Uitto, J. Unco-ordinate regulation
140). of collagenase, stromelysin and TIMP genes by
87. Phillipson, J.D. 50 years of medicinal plant research PGE2: selective enhancement of collagenase gene
– every progress in methodology is a progress in expression in human dermal fibroblasts in culture.
science. Planta Med. 69, 491–495 (2003). J. Cell Biochem. 54, 465–472 (1994).
88. Russo, A., Izzo, A.A., Cardile, V., Borrelli, F. and 103. Varani, J., Fisher, G.J. and Voorhees, J.J. Method for
Vanella, A. Indian medicinal plants as antiradicals protecting and restoring skin using selective MMP

ª 2005 International Journal of Cosmetic Science, 27, 17–34 33


Ultraviolet radiation and skin aging S. Pillai, C. Oresajo and J. Hayward

inhibitors. US Patent application no. 20020119107 rich polysaccharide. Biomed. Pharmacother. 56,
(2002). 258–264 (2002).
104. Pentapharm news release. IFSCC news release from 113. Cha, B.Y., Bpark, C.J., Lee, D.G. et al. Inhibitory
Phenta pharm at http://www.pentapharm.com on effect of methanol extract of Euonymus alatus on
PEPHA-TIMP, a product to inhibit MMP and stimu- MMP-9. J. Ethanopharmacol. 85, 1637 (2003).
late collagen synthesis. 114. Sun, Y. and Xu, Q. Aqueous extract from Rhizoma
105. Hantke, B., Lahmann, C., Venzke, K. et al. Influence notopterygii reduces conteact sensitivity by inhibiting
of flavonoids and vitamins on the MMP- and TIMP- lymphocyte migrationvia down-regulating MMP
expression of human dermal fibroblasts after UVA activity. Pharmacol. Res. 46, 333–337 (2002).
irradiation. Photochem. Photobiol. Sci. 1, 826–833 115. Rothenberg, M.L., Nelson, A.R. and Hande, K.R.
(2002). New drugs on the horizon: matrix metalloprotein-
106. Stab, F. and Tschesche, H. Influence of flavonoids ase inhibitors. Oncologist 3, 271–274 (1998).
and vitamins on the MMP- and TIMP-expression of 116. Holleran, W.M., Galardy, R.E., Gao, W.N. et al.
human dermal fibroblasts after UVA irradiation. Matrix metalloproteinase inhibitors reduce phorbol
Photochem. Photobiol. Sci. 1, 826–833 (2002). ester-induced cutaneous inflammation and hyperpl-
107. Kut-Lasserre, C., Miller, C.C., Ejeil, A.L. et al. Effect asia. Arch. Dermatol. Res. 289, 138–144 (1997).
of avocado and soybean unsaponifiables on gela- 117. Yamamoto, M., Tsujishita, H., Hori, N. et al. Inhibi-
tinase A (MMP-2), stromelysin 1 (MMP-3), and tis- tion of membrane-type 1 matrix metalloproteinase
sue inhibitors of matrix metalloproteinase (TIMP- 1 by hydroxamate inhibitors: an examination of the
and TIMP-2) secretion by human fibroblasts in cul- subsite pocket. J. Med. Chem. 41, 1209–1217
ture. J. Periodontol. 72, 1685–1694 (2001). (1998).
108. Demeule, M., Brossard, M., Page, M., Gingras, 118. Brown, P.D. Ongoing trials with matrix metallopro-
D. and Beliveau, R. Matrix metalloproteinase inhi- teinase inhibitors. Exp. Opin. Investig. Drugs 9,
bition by green tea catechins. Biochim. Biophys. 2167–2177 (2000).
Acta 1478, 51–60 (2000). 119. Ying, Q.L., Rinehart, A.R., Simon, S.R. and Cheron-
109. Morrow, D.M., Fitzsimmons, P.E., Chopra, M. and is, J.C. Inhibition of human leucocyte elastase by
McGlynn, H. Dietary supplementation with the ursolic acid: evidence for a binding site for pentacy-
anti-tumour promoter quercetin: its effects on mat- clic triterpenes. Biochem. J. 277, 521–526 (1991).
rix metalloproteinase gene regulation. Mutat. Res. 120. Sartor, L., Pezzato, E. and Garbisa, S. (-)Epigallo-
1, 480–481 (2001). catechin-3-gallate inhibits leukocyte elastase:
110. Song, L., Xu, M., Lopes-Virella, M.F. and Huang, potential of the phyto-factor in hindering inflamma-
Y. Quercetin inhibits matrix metalloproteinase-1 tion, emphysema, and invasion. J. Leukoc. Biol. 71,
expression in human vascular endothelial cells 73–79 (2002).
through extracellular signal-regulated kinase. Arch. 121. Simon, S., Vered, M., Rinehart, A., Cheronis, J. and
Biochem. Biophys. 391, 72–78 (2001). Janoff, A. Inhibition of human neutrophil elastase
111. Sato, T., Koike, L., Miyata, Y. et al. Inhibnition of by polyguanylic acid and other synthetic polynucle-
AP-1binding activity and PIP-3-kinase pathway by otides. Adv. Exp. Med. Biol. 240, 65–74 (1988).
nobiletin, a polymethoxy flavonoid, results in aug- 122. Tyagi, S. and Simon, S.R. Parinaric acids as probes
mentation of TIMP-1 production and suppression of of binding domains in neutrophil elastase. J. Biol.
production of MMP-1 and -9 in human fibrosarco- Chem. 266, 15185–15191 (1991).
ma HT-1080 cells. Cancer Res. 62, 1025–1029 123. Tyagi, S. and Simon, S.R. Inhibitors directed to
(2002). binding domains in neutrophil elastase. Biochemis-
112. Isnard, N., Peterszegi, G., Robert, A.M. and Robert, try 20, 9970–9977 (1990).
L. Regulation of elastase type endoprptidase activ- 124. Tyagi, S. and Simon, S.R. Interaction of neutrophil
ity, MMP-2 and MMP-9 expression and activation elastase with hydrophobic polyanoinic chelators.
in human dermal fibroblasts by fucose and fucuse- Biochem. Cell Biol. 69, 624–629 (1991b).

34 ª 2005 International Journal of Cosmetic Science, 27, 17–34

You might also like