Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

Ageing Research Reviews 31 (2016) 36–54

Contents lists available at ScienceDirect

Ageing Research Reviews


journal homepage: www.elsevier.com/locate/arr

Skin aging and oxidative stress: Equol’s anti-aging effects via


biochemical and molecular mechanisms
Edwin D. Lephart ∗
Department of Physiology and Developmental Biology and The Neuroscience Center, Brigham Young University, Provo, UT 84602, USA

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

Article history: Oxygen in biology is essential for life. It comes at a cost during normal cellular function, where reactive
Received 2 June 2016 oxygen species (ROS) are generated by oxidative metabolism. Human skin exposed to solar ultra-violet
Received in revised form 29 July 2016 radiation (UVR) dramatically increases ROS production/oxidative stress. It is important to understand
Accepted 4 August 2016
the characteristics of human skin and how chronological (intrinsic) aging and photo-aging (extrinsic
Available online 9 August 2016
aging) occur via the impact of ROS production by cascade signaling pathways. The goal is to oppose
or neutralize ROS insults to maintain good dermal health. Botanicals, as active ingredients, represent
Keywords:
one of the largest categories used in dermatology and cosmeceuticals to combat skin aging. An emerg-
Free radicals
Oxidative stress
ing botanical is equol, a polyphenolic/isoflavonoid molecule found in plants and food products and via
Antioxidant gastrointestinal metabolism from precursor compounds. Introductory sections cover oxygen, free radi-
Anti-aging cals (ROS), oxidative stress, antioxidants, human skin aging, cellular/molecular ROS events in skin, steroid
Equol enzymes/receptors/hormonal actions and genetic factors in aging skin. The main focus of this review cov-
Human skin ers the characteristics of equol (phytoestrogenic, antioxidant and enhancement of extracellular matrix
properties) to reduce skin aging along with its anti-aging skin influences via reducing oxidative stress
cascade events by a variety of biochemical/molecular actions and mechanisms to enhance human dermal
health.
© 2016 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
1.1. Oxygen in biology (Pros and cons) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
1.2. Free radicals (ROS) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
1.3. Oxidative stress . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
1.4. Antioxidants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
2. Human skin characteristics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
3. Aging of human skin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
3.1. Intrinsic (chronological) and extrinsic (photo) aging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
3.2. Cellular and molecular ROS events in human skin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
3.3. Skin steroid enzymes, steroid hormones and steroid receptors in human aging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43
3.4. Genetics and steroid hormonal factors in skin aging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 44
4. Characteristics of equol: chemical structure, isomeric forms, metabolic and plant sources and biological actions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 44
4.1. Antioxidant properties of equol . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
4.2. Equol’s lipophilic nature and penetration into human skin via a reservoir delivery mechanism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
5. Equol’s anti-aging influence on human skin by reducing oxidative stress in cascade events . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46
5.1. Equol decreases ROS via sStimulation of Nrf2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 47

∗ Corresponding author at: Department of Physiology and Developmental Biol-


ogy and The Neuroscience Center, LS 4005, College of Life Sciences, Brigham Young
University, Provo, UT 84602, USA .
E-mail address: Edwin Lephart@byu.edu

http://dx.doi.org/10.1016/j.arr.2016.08.001
1568-1637/© 2016 Elsevier B.V. All rights reserved.
E.D. Lephart / Ageing Research Reviews 31 (2016) 36–54 37

5.2. Equol acts as an antioxidant, stimulates antioxidant/detoxifying enzymes and inhibits skin aging biomarkers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 47
5.3. Equol protects DNA and enhances nerve and tissue repair . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48
5.4. Equol inhibits AP-1 and neoplastic cell growth . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48
5.5. Equol inhibits NFkappaB . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48
5.6. Equol inhibits the inflammatory response . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49
5.7. Equol inhibits MMPs and elastase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49
5.8. Equol stimulates TIMP 1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49
5.9. Equol stimulates collagen . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49
5.10. Equol stimulates elastin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49
6. Equol: comparison to resveratrol . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49
7. Summary and conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 50
Conflict of interests . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51
Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51

1. Introduction steroid enzymes/receptors and hormonal actions are covered along


with the influence of genetic factors in skin aging. Then, the main
Oxidative stress plays an important role in human skin aging sections of this review include the characteristics of equol and
and dermal damage (Gonzaga, 2009; Kammeyer and Luiten, 2015; its anti-aging influence on human skin via reducing oxidative
Natarajan et al., 2014; Rittie and Fisher, 2002; Zouboulis and stress cascade events by a variety of biochemical and molecular
Makrantonaki, 2011). The mechanisms of intrinsic (chronological) actions/mechanisms. Each section is self-contained with brief back-
and extrinsic (photo) aging include the generation of reactive oxy- ground information for each topic followed by examples/figures
gen species (ROS) via oxidative metabolism and exposure to sun showing human applications and/or analysis for the cited studies
ultra-violet (UV) light, respectively (Gonzaga, 2009; Kammeyer and presented.
Luiten, 2015; Natarajan et al., 2014). Photo-aging is dependent
upon the exposure to and intensity of solar UV radiation (UVR) 1.1. Oxygen in biology (Pros and cons)
(Gonzaga, 2009; Kammeyer and Luiten, 2015). Oxidant events and
molecular mechanisms of skin aging involve damage to DNA, the The respiration system in humans provides the breath of life that
inflammatory response, reduced production of antioxidants and is dependent upon oxygen. The oxygen from the atmosphere trans-
the generation of matrix metalloproteinases (MMPs) that degrade ferred into blood is utilized to metabolize dietary nutrients (e.g.,
collagen and elastin in the dermal skin layer (Gonzaga, 2009; fats, proteins & carbohydrates) in order to produce energy. In cellu-
Kammeyer and Luiten, 2015; Natarajan et al., 2014; Rittie and lar aerobic metabolic respiration, glucose is typically the molecule
Fisher, 2002; Zouboulis and Makrantonaki, 2011). All of these that is utilized to generate energy-rich ATP molecules within the
events lead to damaged skin and reflect the aging process: the mitochondria of the cell to maintain homeostasis (Lephart, 2009;
disruption of the extra cellular dermal matrix, the loss of tensile Martini, 2004).
strength and elasticity, impaired wound healing, the appearance of While biological systems are dependent upon atmospheric oxy-
wrinkles, age-spots and loss of skin tone. The goal is to delay aging gen, too little or too much oxygen can be damaging to cells and
onset and/or slow down the structural and visual appearance of tissues (Lephart, 2009; Martini, 2004). In this regard, oxygen is
skin aging with time. often referred to as a Janus gas that has both positive benefits and
Because phytochemicals are known to be constituents of animal potentially damaging side-effects in biological systems (Burton and
and human food sources, botanicals with polyphenolic structures Jauniaux, 2011). Oxygen participates in high-energy electron trans-
have received increased research study due to their potential sig- fers that supports the generation of abundant essential quantities
nificance and application in treating human cancers and other of ATP molecules through oxidative metabolism (Lephart, 2009;
age-related diseases including skin aging (Adlercreutz et al., Martini, 2004). However, biological systems and the numerous
2004; Evans and Johnson, 2010; Lephart et al., 2014; Mazur and chemical reactions within cells are not machines, and errors occur
Adlercreutz, 2000; Park and Pezzuto, 2015; Wang et al., 2014). resulting in the generation of very harmful molecules or particles
Isoflavonoid (plant-derived) molecules that fall under the polyphe- that are associated with oxygen such as free radicals.
nolic umbrella have been shown to decrease oxidative stress
(Bansal and Parle, 2010; Djuric et al., 2001; Yoon and Park, 2014). 1.2. Free radicals (ROS)
One of the important emerging isoflavonoid molecules is equol,
which can decrease oxidative stress (Ma et al., 2010; Zhang et al., While oxygen is essential for life, its benefits have a cost in
2013). In skin, equol has been shown to improve dermal health normal cellular function. As discussed by Halliwell and Gutteridge
by direct and downstream influences at several different steps (1999), unless electrons are transferred to oxygen during aerobic
of the oxidative stress cascade, while at the same time inhibit metabolism in the correct manner, potentially oxygen free radicals
MMP’s actions and simultaneously stimulate collagen and elastin can be formed easily. Occasionally electrons “escape,” and instead
(Gopaul et al., 2012; Lephart, 2013a). Finally, equol has two primary of completing the cellular respiration cycle, oxygen may become
properties: 1) an inherent antioxidative capacity resulting from its toxic and mutagenic (Halliwell and Gutteridge, 1999). For example,
polyphenolic molecular structure and 2) its well-known phytoe- a single oxygen atom is unstable and wants to bind a twin atom,
strogenic activity that enables it to reduce skin aging (Gopaul et al., forming molecular oxygen (O2 ). However, when this is not possible,
2012; Lephart, 2013a). the stability of this bond is compromised because only one pair of
In summary, the general concepts and principles of oxygen electrons is shared and two unpaired electrons remain (Fig. 1). This
in biology (pros and cons), free radicals (ROS), oxidative stress, complex represents a superoxide (O2 − ) and hydroxyl radical. These
antioxidants, human skin [intrinsic and extrinsic (photo)] aging are often referred to collectively as reactive oxygen species (ROS)
and, cellular and molecular ROS events are covered first. Also, due to the presence of an unpaired electron, which makes them
highly reactive (Buonocore et al., 2010; Halliwell and Gutteridge,
38 E.D. Lephart / Ageing Research Reviews 31 (2016) 36–54

1.4. Antioxidants

The body’s two major defense systems are free radical detox-
ifying enzymes and antioxidant molecules (Krishnamurthy and
Wadhwani, 2012; Martini, 2004). In brief, the free radical detoxify-
ing enzyme systems include: superoxide dismutase (SOD), catalase
and glutathione peroxidases. SOD is an enzyme that catalyzes the
partitioning of the superoxide (• O2 − ) into either molecular oxy-
gen (O2 ) or hydrogen peroxide (H2 O2 ) (Fig. 1) (Krishnamurthy
and Wadhwani, 2012). Catalase is another important detoxifying
enzyme that converts hydrogen peroxide to water and molecular
oxygen; it is also inducible and thus completes the action of SOD
(Krishnamurthy and Wadhwani, 2012) (Fig. 1). Glutathione perox-
idase is similar to catalase and represents a family of enzymes that
covert hydrogen peroxide into water and oxygen (Krishnamurthy
and Wadhwani, 2012). Notably, SOD, catalase and glutathione per-
Fig. 1. Electron structures of some common reactive oxygen species (ROS). Each
structure is provided with its name and chemical formula. The red • indicates an
oxidase are important antioxidant enzymes in human skin (Shindo
unpaired electron. Molecular oxygen (O2 ) becomes Superoxide (• O2− ) with the et al., 1994).
loss of an electron. Hydrogen Peroxide (H2 O2 ) becomes Peroxide (• O2−2 ) with the An antioxidant is any molecule that can block free rad-
loss of two electrons. Superoxide Dismutase (SOD) converts Superoxide into either icals and/or ROS from stealing electrons from other atoms
Molecular Oxygen or Hydrogen Peroxide and then Catalase (CAT) converts Hydrogen
(Krishnamurthy and Wadhwani, 2012; Martini, 2004). Or, in other
Peroxide into Water and Molecular Oxygen (e.g., 2H2 O2 → 2H2 O + O2 ).
words, an antioxidant is any substance that inhibits the damage
due to oxygen (oxidation) that is caused by free radicals or reac-
tions promoted by ROS. Antioxidants act at intra- and extra-cellular
locations (Ames et al., 1981; Becker, 1993; Krishnamurthy and
1999) and capable of chain reactions, which form another free rad-
Wadhwani, 2012; Martini, 2004). Glutathione and uric acid are two
ical at each step (Buonocore et al., 2010; Halliwell and Gutteridge,
molecules the body can synthesize that are endogenous sources
1999; Kohen and Nyska, 2002).
of antioxidants (Krishnamurthy and Wadhwani, 2012). Both glu-
The overproduction of ROS can have deleterious effects on cell
tathione and uric acid are important antioxidants present in human
structures, including damage to membranes, lipids, proteins, RNA
skin (Shindo et al., 1994).
and DNA (Buonocore et al., 2010; Gonzaga, 2009; Halliwell and
Many different antioxidants are obtained from dietary or exoge-
Gutteridge, 1999; Kammeyer and Luiten, 2015; Kohen and Nyska,
nous sources. The most commonly known antioxidants include,
2002; Natarajan et al., 2014; Rittie and Fisher, 2002; Zouboulis
vitamins A, C and E (Krishnamurthy and Wadhwani, 2012;
and Makrantonaki, 2011). In this regard, ROS production leads to
Martini, 2004). However, other dietary sources of antioxidants are
the mechanisms of oxidative stress in living systems that account
carotenoids, lipoic acid and phenolic compounds found in abun-
for a majority of disorders, diseases and death worldwide. Finally,
dance in many plant products (Krishnamurthy and Wadhwani,
ROS are well known to be involved in human cutaneous aging, the
2012; Shindo et al., 1994). Many of these dietary antioxidants are
dermal inflammatory response and skin cancers (Gonzaga, 2009;
incorporated into the epidermal and dermal layers of human skin
Kammeyer and Luiten, 2015; Natarajan et al., 2014; Rittie and
(Shindo et al., 1994).
Fisher, 2002; Zouboulis and Makrantonaki, 2011).
Finally, exposure to solar UVR, heavy metals or other toxic
agents are known to increase free radical or ROS formation along
with the inescapable generation of ROS by normal metabolic
1.3. Oxidative stress oxidation associated with mitochrondrial function (Tulah and
Birch-Machin, 2013). Thus, a healthy life free of disease becomes
Kohen and Nyska (2002) noted that ROS are involved in a a matter of balance; biological systems must have enough antiox-
variety of biological phenomena, such as mutation, carcinogene- idants available that are ready to “neutralize” various free radicals
sis, degeneration and other disorders and disease states involving or ROS the body is either exposed to or that it produces. If an imbal-
inflammation and aging. All major organ systems in humans includ- ance occurs, then oxidative stress will result and homeostasis will
ing skin, nervous, cardiovascular, respiratory, kidney, skeletal and not be maintained.
immune, etc., along with other multiple organs disorders such as
diabetes, sepsis, depression and trauma are impacted by oxida-
tive stress (Bar-Or et al., 2015; Kohen and Nyska, 2002; Maritim
et al., 2003; Maes et al., 2011). Thus, based upon numerous jour- 2. Human skin characteristics
nal reports, ROS generation represents the root basis of oxidative
stress that leads to oxidative damage in biological structures and Human skin is the largest and most complex organ (represent-
molecules (Bar-Or et al., 2015; Gonzaga, 2009; Kammeyer and ing one sixth of the total body weight) functioning as a physical
Luiten, 2015; Maes et al., 2011; Maritim et al., 2003; Natarajan barrier to protect the body from water loss as well as environ-
et al., 2014; Rittie and Fisher, 2002; Valko et al., 2007; Zouboulis and mental insults such as pathogens, chemicals, physical agents and
Makrantonaki, 2011). Oxidative stress may be defined as the imbal- solar UVR throughout life (Madison, 2003; Qunshan and Nash,
ance between the production of free radicals and the ability of the 2010; Shindo et al., 1994). Moreover, the skin provides essential
body to counteract or detoxify their harmful effects through neu- physiological functions including immune defense, thermoregu-
tralization by antioxidants. Since free radical (ROS) development lation, sensory input from mechanoreceptors and endocrine and
is unavoidable, the human body has adapted by establishing and metabolic mechanisms to sustain optimal health (Lephart, 2009;
maintaining defense mechanisms that reduce their impact, partic- Martini, 2004; Qunshan and Nash, 2010). Finally, and most impor-
ularly in human skin (Kammeyer and Luiten, 2015; Natarajan et al., tantly, the antioxidant defense capacity of the skin would be
2014; Shindo et al., 1994; Zouboulis and Makrantonaki, 2011). expected to be greater than that of internal organs due to its pro-
E.D. Lephart / Ageing Research Reviews 31 (2016) 36–54 39

Fig. 3. Production of estrogen, collagen/elastin and elasticity profiles/patterns in


women with age. Estrogen levels peak in the late twenties, while collagen and elastin
peak around age 30. Elasticity (mechanical “bounce back or recoil”) is greatest in
very young individuals (e.g., at 10 years of age), which continually decline with age.
Profiles were generated via composite data from references (Escoffier et al., 1989;
Hillebrand, 2010; Jain et al., 2003; Schwartz and Mayaux, 1982; Seite et al., 2006;
Uitto, 2008).
Fig. 2. A cartoon graphic displaying the three skin layers in humans. Keratinocytes
are the major cellular type in the epidermis. Melanocytes near the epidermal-dermal
junction provide pigmentation of the skin for photo-protection. In the dermis, the supportive glycosaminoglycans that maintain optimal skin health
blue horizontal bars represent collagen fibers while the green vertical bars indicate
(Farage et al., 2010; Lephart, 2009; Martini, 2004). It is composed
elastin fibers.
mainly of extracellular matrix proteins (such as collagen, elastin,
etc.) that are secreted by fibroblasts and provide dermal strength
tective structural and biological functioning of the dermal layers and flexibility (Farage et al., 2010; Lephart, 2009; Martini, 2004;
(Shindo et al., 1994). Qunshan and Nash, 2010). The elastic fiber network is responsible
The skin is divided into three distinct layers: 1) the epidermis; 2) for recoil and elasticity of the skin, but it also plays a role in tis-
the dermis; and 3) the hypodermis or subcutaneous tissue (Lephart, sue repair (Lephart, 2009; Pierard et al., 2010; Mera et al., 1987)
2009; Madison, 2003; Martini, 2004; Qunshan and Nash, 2010). (Fig. 2). The dermal layer also contains other molecules such as
See Fig. 2 for a basic graphic representation of human skin layers. tissue inhibitor of matrix metalloproteinases (TIMP), metallopro-
However, in brief, only the epidermis and dermis will be covered teinases (MMPs), elastase and many other molecules that maintain
due to their structural/functional components in reference to skin skin health (Farage et al., 2010).
characteristics and aging.
The epidermis is the major protective outer layer. The stra- 3. Aging of human skin
tum corneum (10–30 ␮m) is the outermost layer of progressively
dying and flattened dead cells or corneocytes. The ‘bricks and mor- Aging is accompanied by the progressive loss of anatomical
tar’ cellular structure is composed of the live keratinocyte layers, structure and physiological function in multiple organs. In 2050,
which form the majority of the epidermal layer (100–150 ␮m) the U. S. population aged 65 and over is projected to be 83.7 mil-
and its active functional aspects (Farage et al., 2010; Madison, lion, almost double its estimated population of 43.1 million in 2012
2003; Menon, 2002; Qunshan and Nash, 2010; Shindo et al., 1994). (Ortman et al., 2014). In Western countries it is estimated that
Melanocytes near the epidermal-dermal junction provide pigmen- women will spend more than one-third of their lifetimes in post
tation of the skin for photoprotection (Natarajan et al., 2014) (Fig. 2). menopause and more than 40 million postmenopausal women live
Conspicuously, the epidermis, composed mainly of ker- in the U.S. today composing approximately 18% of the total popu-
atinocytes, has a greater abundance of the antioxidant enzymes lation (Brincat et al., 2005; Farage et al., 2008).
such as SOD (125%), catalase (720%) and glutathione peroxidase As human beings age, the skin thins, dries, wrinkles, becomes
(60%) compared to the dermal layer (Shindo et al., 1994). Also, pigmented unevenly with age or liver spots (solar lentigines), and
seasonal variations (summer vs. winter) have been reported for wound healing is delayed (Friedman, 2005; Pierard et al., 2010).
catalase, while SOD enzyme activity remains stable, when human Specifically, the appearance of wrinkles around the eyes and mouth,
skin is exposed to UV light (Hellemans et al., 2003). and frown lines along the forehead are seen with uneven skin color
Additionally, the biosynthesized antioxidants glutathione and and a general loss of skin tone (pale appearance). Sagging skin and
uric acid, along with the dietary-derived antioxidants, vitamin D thin skin are due to the loss of and definition/abundance of the
and vitamin E (tocopherols), are present at much higher concentra- underlying collagen and especially the elastin fibers in the dermal
tions in the epidermis compared to the dermal layer (Shindo et al., layer that provide full, robust and the elastic recoil properties of
1994). Thus, the epidermal layer contains the highest concentration youthful skin (Duncan and Leffell, 1997).
of antioxidants and is the major line of antioxidant defense in skin Skin collagen and elastin peaks around 30 years of age (Burns
(Natarajan et al., 2014; Seo and Chung, 2006; Shindo et al., 1994). and Breathnach, 2004; Fazio et al., 1988; Freedberg et al., 2003; Khol
While the epidermis provides the first line of defense, the der- et al., 2011; Seite et al., 2006), which corresponds with the peak of
mis bestows the scaffolding or structural fibers of the skin. In estrogen production around 25–30 years of age (Baumann, 2002;
general, the dermis is a dense and irregular layer of connective Jain et al., 2003; Schwartz and Mayaux, 1982) (Fig. 3). Reported fea-
tissue, 2–3 mm thick, that comprises most of the skin thickness tures of aged human skin include fragmentation of collagen fibers
(Brincat et al., 2005). It contains mechanosensory receptors, sweat by the action of the enzyme MMP-1 and increased mitochondrial
and oil (sebaceous) glands, and its primary water-holding com- ROS production and oxidative stress resulting in common dele-
ponents i.e., hyaluronic acid (responsible for normal turgor) and tions of mitochondrial DNA (Ashworth et al., 1999; Qin et al., 2014;
40 E.D. Lephart / Ageing Research Reviews 31 (2016) 36–54

Quan et al., 2015). Also, the importance of SOD in skin aging has by the ozone layer and do not reach the surface of the earth
been shown in SOD-deficient mice where epidermal thinning was (Gonzaga, 2009; Walker et al., 2003). UVA and UVB make up 95–98%
induced by DNA breaks causing cellular senescence (Velarde et al., and 2–5%, respectively, of the UV radiation reaching human skin
2012). (Walker et al., 2003). However, the precise amount and type of UVR
The first signs of skin aging occur around age 30 and especially (especially UVB) exposure depends on a number of factors (solar
after menopause, where there is skin dryness, decreased skin firm- zenith angle, latitude; stratospheric ozone levels, and pollution,
ness and loss of elasticity (Burns and Breathnach, 2004; Escoffier weather-cloud cover, and altitude) (Walker et al., 2003) (Fig. 5).
et al., 1989; Fazio et al., 1988; Freedberg et al., 2003; Hillebrand, UVB (290–320 nm) represent only 2–5% of the sun’s emissions,
2010; Khol et al., 2011; Seite et al., 2006; Uitto, 2008). In fact, elas- it penetrates into epidermal cells, can damage DNA and activate a
ticity has been shown (via mechanical depression) to be highest cascade of events leading to photo-aging (Walker et al., 2003). UVA
in young (10-year-old) subjects with a continual gradual decline (400–320 nm) represents 95–98% of the total UV radiation reach-
with chronological aging (Uitto, 2008) (Fig. 3). While skin aging ing earth’s surface (Walker et al., 2003). While also damaging the
first appears around 30 years of age, an important study has clearly epidermis, UVA penetrates deeper into the dermis to degrade col-
demonstrated that the age-related loss in skin elasticity actually lagen and elastin fibers via oxidative stress and activating MMPs,
precedes the formation of visible wrinkles (Fujimura et al., 2007). thus, UVA is more cytotoxic than UVB (Gonzaga, 2009; Kammeyer
Behind collagen, the importance of elastin is paramount, since and Luiten, 2015; Natarajan et al., 2014; Rittie and Fisher, 2002;
loss of elastin can result in rapid skin aging (Farage et al., 2010; Zouboulis and Makrantonaki, 2011). The characteristics of light
Qunshan and Nash, 2010). As discussed by Anderson (2012) when and UV light (UVC, UVB and UVA) penetration into human skin are
a 21-year-old women experienced an allergic food reaction, and all summarized in Fig. 5.
elastin was selectively destroyed that resulted in rapid skin damage In sun-exposed skin areas, such as the face and neck, photo-
and aging over a period of a few years (Fig. 4). Remarkably, as dis- aging pathological changes in cellular activities occur with the
cussed by Anderson, there is not a clear line of distinction between generation of ROS that lead to the gross disorganization of the
medical procedures in dermatology and cosmetic applications to dermal matrix and the decline of antioxidants (Gonzaga, 2009;
assist patients to feel better by having a positive perception about Kammeyer and Luiten, 2015; Khol et al., 2011; Natarajan et al.,
themselves (Anderson, 2012; Kligman and Koblenzer, 1997). For 2014; Rittie and Fisher, 2002; Shindo et al., 1994; Zouboulis and
example, whether a person goes through dermal trauma from war Makrantonaki, 2011). How much sun exposure is required to cause
or accidental means compared to individuals experiencing chrono- photo-aging depends on a person’s type of skin (fair vs. darker
logical skin aging, the impact on patients in regard to positive pigmentation; as classified by Fitzpatrick’s skin types) and the
self-perception is paramount especially if the events/changes occur exposure over time without skin protection to UVR. One study
in highly visible body areas such as the face, neck, hands and arm suggested that minimal repetitive exposure to UVR, equivalent
regions (Anderson, 2012; Kligman and Koblenzer, 1997). to 5–15 min of mid-day sun every-other-day was sufficient to
maintain elevated levels of MMPs that are known to degrade der-
3.1. Intrinsic (chronological) and extrinsic (photo) aging mal collagen and elastin fibers (Rabe et al., 2006). Surprisingly,
this is approximately the same amount of sun exposure internal
Skin aging can be classified by intrinsic and extrinsic mech- medicine/endocrinologists suggest is needed to maintain healthy
anisms (Gonzaga, 2009; Farage et al., 2010; Farage et al., 2008; vitamin D levels. (Gilchrest, 2008). In 1998, previous to the above
Kammeyer and Luiten, 2015; Khol et al., 2011; Zouboulis and data, the American Academy of Dermatology issued the “safe sun
Makrantonaki, 2011). Intrinsic or chronological aging is an unavoid- position” based upon the irrefutable facts that UV irradiation causes
able phenomenon that includes several factors such as genetics, skin cancer, melanoma and photo-aging and that vitamin D can be
metabolism and the passage of time, where the “repair process” obtained from the diet or from oral supplements (Robinson et al.,
may become defective (Anderson et al., 2014; Meadows et al., 2014; 1998). However, controversy about dietary vitamin D increasing
Quan et al., 2015; Tulah and Birch-Machin, 2013; Velarde et al., vitamin D status remains a controversy due to: 1) a suggested high
2012). The repair process is an important component with aging, prevalence of low vitamin D intakes and vitamin D deficiency or
since DNA and their gene products (i.e., proteins) play such an inadequate vitamin D status in Europe and 2) comparing vitamin
essential role in optimal dermal health. The generation of mito- D intakes estimated from foods and dietary supplements to serum
chondrial ROS by oxidative metabolism represents the inescapable 25-hydroxy vitamin D concentrations is problematic since com-
production of free radicals by aerobic cellular chemical reactions in parisons can only be made on group means rather than on data
chronological aging leading to mitochondrial damage, altered cell linked to individuals (Spiro and Buttriss, 2014; National Institutes
function and, if the insult is too great, the affected cells may die of Health, Vitamin D, 2016).
(Anderson et al., 2014; Meadows et al., 2014; Quan et al., 2015; Thus, staying out of the sun is the best avoidable practice a per-
Tulah and Birch-Machin, 2013; Velarde et al., 2012). If damaged son can do to prevent ROS production and photo-aging. To illustrate
skin cells are not repaired, mutations occur that result in premature this point, Fig. 6 shows a 69-year-old man that drove trucks com-
aging (Qin et al., 2014; Quan et al., 2015). mercially for twenty-eight years. As Fig. 6 displays, the left side of
On the other hand, extrinsic aging is a phenomenon that can be this man’s face (driver’s side) shows dramatic photo-aging, while
avoided to some extent. Extrinsic aging is caused by environmental the right side of the face (protected from UVR), shows minimal
exposure, primarily solar UV radiation (UVR) or ultra violet (UV) photo-aging (Gordon and Brieva, 2012). This condition is unilateral
light from artificial tanning sources. This is more commonly termed dermatheliosis and demonstrates the dramatic impact photo-aging
photo-aging. Photo-aging is the effect of long-term UV exposure has on dermal health.
and the resulting solar damage superimposed on intrinsically aged
skin (Gonzaga, 2009; Kammeyer and Luiten, 2015; Zouboulis and 3.2. Cellular and molecular ROS events in human skin
Makrantonaki, 2014). It is thought that up to 80-to-90% of skin aging
is due to the deleterious effects of the sun or photo-aging (Gonzaga, Recall the unavoidable component of skin aging is chrono-
2009; Hillebrand, 2010; Kammeyer and Luiten, 2005; Khol et al., logical or intrinsic aging (Gonzaga, 2009; Kammeyer and Luiten,
2011; Natarajan et al., 2014). 2015; Natarajan et al., 2014; Rittie and Fisher, 2002; Zouboulis and
There are different types of solar UV light rays (UVC, UVB and Makrantonaki, 2011). In intrinsic aging there is ROS production
UVA) pertaining to human skin photo-aging. UVC rays are blocked via oxidative metabolism (Anderson et al., 2014; Gonzaga, 2009;
E.D. Lephart / Ageing Research Reviews 31 (2016) 36–54 41

Fig. 4. A 21 year-old women (left panel) experienced an allergic food reaction, and all elastin was selectively destroyed resulting in rapid skin damage and aging over a
period of a few years (right panel at age 26) as reported by Anderson (2012).
Photo Source: http://www.telegraph.co.uk/news/newstopics/howaboutthat/8826277/Mystery-condition-makes-woman-age-50-years-in-just-a-few-days.html, October
14, 2011.

Fig. 5. The solar radiation spectrum and the influences of ultra violet (UV) light on skin aging. Brief descriptions of UVC, UVB and UVA characteristics are shown especially
in reference to specific amounts and patterns of UV penetration into the skin and the potential damage each can cause with exposure. For instance, UVC rays do not reach
the surface of the earth. They are blocked by the ozone layer of the earth’s atmosphere (Walker et al., 2003). UVA and UVB make up 95–98% and 2–5%, respectively, of the
UV radiation reaching human skin (Walker et al., 2003). UVB rays penetrated into the epidermal cells can damage DNA and activate the ROS cascade of events leading to
photo-aging. UVA rays while also damaging the epidermis penetrate deeper into the dermis to degrade collagen and elastin fibers via oxidative stress and activating MMPs.
UVA is more cytotoxic in skin than UVB rays.

Meadows et al., 2014). All of the skin aging characteristics (covered important for optimal skin health, this will be a major focus for the
above) are associated with oxidative metabolism and subsequent cellular, molecular, enzymatic, antioxidant and other components
ROS generation that define this inevitable phenomenon. Since the that regulate these extracellular matrix proteins.
structural and functional aspects of collagen and elastin fibers are so
42 E.D. Lephart / Ageing Research Reviews 31 (2016) 36–54

Zouboulis and Makrantonaki, 2011). The activation of the inflam-


matory response generates cytokines or interleukins (ILs), which
then in a positive feedback loop, stimulates the further produc-
tion of ROS (Gonzaga, 2009; Kammeyer and Luiten, 2015; Naylor
et al., 2011; Peng et al., 2015; Rittie and Fisher, 2002; Zouboulis and
Makrantonaki, 2011). These pathways are shown in Fig. 7.
Now, turning to photo- or extrinsic aging, the exposure to UVR
displays the same molecular to cellular responses as for intrin-
sic aging (covered above), but the magnitude of the effects is
amplified, which explains why photo-aging accounts for approx-
imately 80-to-90% of skin aging. It is known that UVR exposure and
ROS production in photo-aging cause damage to DNA, proteins,
lipids and reduces the levels of antioxidants in the skin (Bickers
and Athar, 2006; Gilchrest, 1989; Gonzaga, 2009; Kammeyer and
Luiten, 2015; Natarajan et al., 2014; Naylor et al., 2011; Peng et al.,
2015; Rittie and Fisher, 2002; Shindo et al., 1994; Thiele et al.,
1998; Warren et al., 1991; Zouboulis and Makrantonaki, 2011)
Fig. 6. Photograph of a 69-year-old man that drove commercial trucks for 28 years
(Fig. 7). For instance, vitamin E sequestered in hydrophobic lipids,
that displays the effects of extrinsic or photo-aging. The left-side of this man’s face
(driver’s side) shows dramatic photo-aging, while the right side of his face (pro- can absorb the energy from UV light, and thus plays an impor-
tected from UVR), shows minimal photo-aging. This is a clear example of unilateral tant role in photo-protection from ROS (Rhie et al., 2001; Weber
dermatheliosis from photo-aging. Reproduced with permission from Massachusetts et al., 1997). However, exposure to UV light lowers vitamin E con-
Medical Society License to E.D. Lephart (Gordon and Brieva, 2012). tained primarily in the stratum corneum (Shindo et al., 1994),
which is an early and sensitive in vivo marker of UV induced photo-
oxidation (Thiele et al., 1998). Also, vitamin E concentrations in the
From molecular to cellular cascades, ROS production is known human epidermis decline with age (Rhie et al., 2001). However,
to cause the activation of activator protein-1 (AP-1), which sup- at the same time, these events are also associated with stimula-
presses TGF␤ receptors that in turn blocks pro-collagen synthesis tion of MMPs and a concept called “fibroblast collapse” (Gonzaga,
that reduce collagen levels (Gonzaga, 2009; Kammeyer and Luiten, 2009; Kammeyer and Luiten, 2015; Khol et al., 2011; Natarajan
2015; Naylor et al., 2011; Peng et al., 2015; Rittie and Fisher, 2002; et al., 2014; Rittie and Fisher, 2002; Zouboulis and Makrantonaki,
Zouboulis and Makrantonaki, 2011). At the same time, AP-1 acti- 2011). In this situation fibroblasts increase the production of elas-
vation: a) stimulates MMPs that degrade collagen and b) activates tase, which degrades elastin that in turn reduce elastin levels
NF-kappaB, which is a major activator of the inflammatory response (Akhtar et al., 2010; Gonzaga, 2009; Hillebrand, 2010; Kammeyer
(Gonzaga, 2009; Kammeyer and Luiten, 2015; Natarajan et al., and Luiten, 2015; Khol et al., 2011; Natarajan et al., 2014; Rittie
2014; Naylor et al., 2011; Peng et al., 2015; Rittie and Fisher, 2002;

Fig. 7. Chronological aging via oxidative metabolism and photo-aging by exposure to UV light (extrinsic aging) through cellular/molecular signaling mechanisms is shown.
The cascade events including the major impact of oxidative stress via the generation of reactive oxygen species (ROS) is displayed in reference to the appearance of damaged
skin and wrinkles due to changes in human dermal structural proteins (collagen and elastin). Pro-inflammatory Transcription Factor NF-kappB (NFkappaB), AP-1 is a nuclear
transcription element, Activator Protein − 1 (AP-1) and Transforming Growth Factor beta (TGF␤).
E.D. Lephart / Ageing Research Reviews 31 (2016) 36–54 43

Fig. 8. Steroid hormone enzymes, steroid hormones and steroid receptors in the epidermal and dermal layers during aging. Estrogen receptor beta (ER ␤) is the predominant
estrogen receptor subtype in human skin. The photomicrograph displays ER ␤ (rust-colored) staining in the keratinocytes in the epidermis and fibroblasts in the dermis.
The aromatase enzyme is also present in dermal fibroblasts that convert androgens and estrogens (not shown). Also, the 5␣-reductase type I enzyme in fibroblasts converts
testosterone (T) into 5␣-dihydrotestosterone (DHT), which in turn binds to the androgen receptor (AR) that has negative effects on skin. For example, androgen hormonal
actions via the AR are known to stimulate matrix metalloproteinases (MMPs) and decrease wound healing. Whereas, estrogenic hormonal actions via ER ␤ are known to
decrease MMPs and enhance wound healing. Notably, ER ␣ expression in human skin is expressed at lower levels compared to ER ␤ expression (not shown). While ER ␣
expression does not change with menopause, ER ␤ expression declines by approximately 15–20%, thus decreasing the positive action of this estrogen receptor subtype for
skin health. Information via composite data from references (Brincat et al., 2005; Gopaul et al., 2012; Inoue et al., 2011; Lephart 2013a, 2015; Makrantonaki and Zouboulis,
2009; Nitsch et al., 2004; Pelletier and Ren, 2004; Pomari et al., 2015; Thornton et al., 2003).

and Fisher, 2002; Zouboulis and Makrantonaki, 2011). In total, the (Pelletier and Ren, 2004; Thornton et al., 2003). Androgen receptors
destructive actions associated with photo-aging are noticeable ini- (AR) are expressed at lower levels compared to ER ␤ in fibrob-
tially by wrinkles followed by skin damage that, if not treated, are lasts in the dermis (Pelletier and Ren, 2004; Thornton et al., 2003).
irreversible. The intracrine sex steroid synthesis and signaling in human epi-
It is beyond the scope of this review to detail, especially the dermal keratinocytes and dermal fibroblasts have been recently
molecular factors, elements and pathways involved in the cascade reported by Pomari et al. (2015) that includes the expression of
with photo-aging and ROS production, assuredly, this information the G-protein-coupled estrogen receptor (GPER1) that apparently
is discussed elsewhere (Bickers and Athar, 2006; Gonzaga, 2009; is involved in estrogen signaling for both dermal cell types.
Kammeyer and Luiten, 2015; Natarajan et al., 2014; Naylor et al., The ER ␤ and androgen receptors play important roles in
2011; Peng et al., 2015; Rittie and Fisher, 2002; Zouboulis and skin and hair health. In general, activation of ER ␤ enhances,
Makrantonaki, 2011). whereas, activation of ER ␣ or hormonal actions via andro-
gen receptors decreases skin, hair and prostate health (Lephart,
2014a). For example, Widyarini et al. (2006a) demonstrated
3.3. Skin steroid enzymes, steroid hormones and steroid receptors the mechanism by which estrogen receptor signaling protected
in human aging against solar-stimulated UV radiation-induced immune suppres-
sion. Markiewicz et al. in (2013), using knock-out mice showed that
It is well known that estrogen improves skin quality and der- ER ␤ meditated skin dermal thickness/collagen deposition occur in
mal health, especially in postmenopausal women by increasing a unique manner. This has implications for the protective effect
skin collagen, elastin deposition (elasticity) and hydration (Archer, of estrogen against exposure to UV light or photo-aging. Also, as
2012; Brincat et al., 2005; Freedberg et al., 2003; Friedman, 2005; discussed by Chang et al. in (2010) ER ␤ activation blocked photo-
Hillebrand, 2010; Khol et al., 2011; Wend et al., 2012). It is also aging (via inhibiting various inflammatory biomarkers such as IL- 1,
known that: a) estrogen suppresses ROS production (Ramara et al., IL-6 and NFkappaB) using selective estrogen receptor compounds.
2007) and b) antioxidant enzyme expression is stimulated by estro- The importance of ER ␤ activation and its beneficial influence in
gen via the ERK1 and ERK2[MAP]/NFkappaB cascade (Borras et al., human skin has been recently reviewed (Jackson et al., 2011). More-
2005). The ovary and skin fibroblasts are capable of synthesiz- over, estrogen related receptor (ERR) gamma (␥) has been identified
ing estrogen from steroid precursors by the aromatase enzyme, in keratinocytoes and fibroblasts in human skin that has positive
but ovarian production dramatically decreases after 30 years of influences on dermal health. For instance, when ERR ␥ is activated
age and especially after menopause (Inoue et al., 2011; Jain et al., in skin, it is thought to protect against neoplastic growth (Krahn-
2003; Schwartz and Mayaux, 1982). Topical moisturizers with Bertil et al., 2008).
science-based active ingredients and/or estrogen or oral hormone Also, the 5␣-reductase enzyme is present in fibroblasts. This
replacement therapy (ERT/HRT) have been shown to improve skin enzyme converts testosterone to the more potent androgen, 5␣-
aging parameters and reverse the negative effects of skin aging dihydrotestosterone (5␣-DHT) that decreases skin fibroblast cell
compared to untreated skin (Archer, 2012; Brincat et al., 2005; viability and wound healing (Gopaul et al., 2012; Makrantonaki and
Freedberg et al., 2003; Hillebrand, 2010; Wend et al., 2012). Zouboulis, 2009; Nitsch et al., 2004) (Fig. 8). In reference to ROS pro-
The most important steroid receptors in skin are estrogen and duction, in general, androgens are known to increase free radical
androgen receptors. Estrogen receptors (ER) are expressed as sub- production from data obtained in cardiovascular studies (Chignalia
types, estrogen receptor alpha (ER ␣) and estrogen receptor beta et al., 2012; Tostes et al., 2016).
(ER ␤; Fig. 8). The predominant subtype of ER in skin and scalp
hair is ER ␤, it is also in keratinocytes, fibroblasts and the hair bulb
44 E.D. Lephart / Ageing Research Reviews 31 (2016) 36–54

botanical that addresses skin aging by decreasing ROS via hor-


monal and molecular mechanisms to provide good dermal health
is presented below.

4. Characteristics of equol: chemical structure, isomeric


forms, metabolic and plant sources and biological actions

Traditionally, equol is classified as a polyphenolic compound


that is a metabolite of daidzein (an isoflavone found in plant and
food products) (Lephart, 2013b; Setchell and Clerici, 2010). Phe-
nolics are a group of compounds having at least one hydroxyl
group attached to an aromatic ring. The most high-profile polyphe-
nolic molecule known to the general public is resveratrol that
equol is structurally related (Lephart et al., 2014; Park and Pezzuto,
2015). Interestingly, genistein, another polyphenolic/isoflavonoid
molecule, was highly studied in the 1980s to the mid-1990s until
the equol hypothesis was proposed (Setchell and Clerici, 2010).
Fig. 9. Chronological and photo-aging is shown by split-face photographs of a 23- Equol, an isoflavonoid, has two phenolic rings with hydroxyl
year-old Caucasian women (left panel) compared to the same women at 61 years
groups on each ring that provide functional points for biological
old (right panel). The right panel photograph shows: sagging jaw line, changes in
lip thickness and cheek fullness, skin texture, the dermal expression displays solar activity (Fig. 10A) (Lephart, 2013b; Lephart, 2015; Setchell and
elastosis and wrinkles with dull skin color and uneven skin tone. Reproduced with Clerici, 2010). While endogenous estrogenic hormones such as
permission from MedSkin (Nkengne and Bertin, 2012). 17␤-estradiol are steroids with a cyclo-hexane-phenantrene par-
ent chemical structure that is derived from cholesterol, equol is not
a steroid. However, equol and 17␤-estradiol have similar chemi-
3.4. Genetics and steroid hormonal factors in skin aging cal structures/confirmations and molecular weights (C15 H14 O3 vs.
C18 H24 O2 ; 242.3 g/mol vs. 272.4 g/mol, respectively) (Fig. 10A and
Image analysis were performed examining monozygotic (“iden- B).
tical”) twins to determine how genetics influences or modulate Equol has a chiral center at carbon 3, and thus can exist in two
photo-aging factors skin patterns and skin damage. Since each mirror image forms known as enantiomers (S-equol and R-equol)
twin pair has an identical chromosomal DNA sequence and since (Lephart, 2013b; Setchell and Clerici, 2010). Racemic equol refers
most twins are raised in the same households and exposed to to exact equal portions of S-equol and R-equol (Lephart, 2013b).
similar environments, similar severities of skin damage/wrinkling However, in man and animals only S-equol is produced by intestinal
between the twins were expected. This is exactly what was found bacteria conversion from daidzein, and some individuals are capa-
(Hillebrand, 2010). The implications from these findings suggested ble of producing higher levels of equol than others (Lephart, 2013b;
that ROS generation and oxidative stress impact was similar in Setchell and Clerici, 2010; Setchell et al., 2005). These individuals
monozygotic twins. have been identified as “equol producer” (Lephart, 2013b; Setchell
Next, to track the progression of photo-skin damage superim- and Clerici, 2010). The term “equol producer” is a descriptive or
posed upon chronological aging an 8-year study was performed arbitrary term for humans that maintain S-equol levels around or
(Hillebrand, 2010). Each subject’s pattern of wrinkling around the above 10–20 ng/ml after consumption of soy food products that
eyes and on the cheek at baseline was compared to the pattern infer protective health benefits (Lephart, 2013b). S-Equol has also
observed after 8 years. The study found that subjects, who were been found in plant products such as beans, cabbage, lettuces, tofu
in their forties at baseline, displayed a significantly faster rate and other food and animal products (Abiru et al., 2012; Common
of skin damage over the 8-year period compared to women in and Ainsworth, 1961; Hounsome et al., 2009, 2010; Jou et al., 2013).
other age groups at baseline, confirming previous reports (Akazaki For example, S-equol has been reported in cow’s milk (Bannwart
et al., 2002; Kuwazuru et al., 2008). Finally, the relationship of et al., 1986; Hoikkälä et al., 2007; King et al., 1998; Lundh et al.,
menopausal status and rate of skin damage was examined. Women, 1990), and Frankenfeld (2011) showed that dairy consumption sig-
who had entered menopause between baseline and 8 years, showed nificantly correlated with urinary equol levels in U.S. adults. Also,
the highest rate of skin damage (>95% increase), suggesting that there are some data that showed R-equol levels in fermented soy
hormone replacement therapy (HRT) is most effective within 5 products are higher compared to S-equol (Lephart, 2013b; Lephart,
years of menopausal (Archer, 2012; Phillips et al., 2008). 2014b). Notably, the metabolism of R- and S-equol in humans
Thus aging can be described as a fast moving train without any appears to be similar (Setchell et al., 2009). Therefore, humans are
stops along the way. Skin aging, along with prostate health changes exposed to this polyphenolic/isoflavonoid compound from differ-
(Lephart, 2014a) and male-pattern baldness (Lee et al., 2015), are ent plant and food sources regardless of age, gender or geographical
associated with intrinsic or chronological alterations such as hor- location with scientific data to support a consumption/exposure
monal changes and for skin exposure to the sun or photo-aging. record that appears to be safe (Andres et al., 2012; Badger et al.,
These aging processes are demonstrated in Fig. 9 showing a woman 2009; Degen et al., 2011; Gilchrist et al., 2010; Hamilton-Reeves
(in a split-face photograph) at 23-years-old vs. 63-years of age et al., 2010).
(Nkengne and Bertin, 2012). The split-face comparison displays Of particular interest from a chemical messenger and molec-
decreased collagen and elastin deposition, increased wrinkle for- ular perspective, equol has an affinity for estrogen receptor beta
mation and dull skin tone. (ER ␤), which is abundant in keratinocytes of the epidermis and
Therefore, the goal to slow down the fast moving train of skin fibroblasts in the dermis (Pelletier and Ren, 2004; Setchell et al.,
aging needs to include lifestyle changes (decreased sun exposure) 2005; Thornton et al., 2003). More recent data suggest that G-
and treatments (decreased ROS production), in order to delay the protein-coupled estrogen receptors (GPER1) are expressed in both
onset/development of damaged cutaneous elements in order to keratinocytes and fibroblasts that mediate estrogenic signaling
support good dermal health. Evidence that equol is an emerging (Pomari et al., 2015). In this regard, there is no evidence, to date,
E.D. Lephart / Ageing Research Reviews 31 (2016) 36–54 45

Fig. 10. Chemical structures, formulas and molecular weights (MW) of Equol (panel A) and of 17␤-Estradiol (panel B). The 2-dimensional chemical structures are shown in
the top half of the figure, while the bottom half displays the 3-dimensional chemical structures of Equol and 17␤-Estradiol (PubChem). While both Equol and 17␤-Estradiol
contain aromatic rings, 17␤-Estradiol is a steroid hormone while Equol is not. Also, Equol has a chiral center at carbon 3 (see blue circle, panel A), and thus can exist in two
mirror image forms known as enantiomers (i.e., S-equol and R-equol). Racemic equol refers to exact equal portions of S-equol and R-equol (Lephart, 2013b). The red spheres
indicate oxygen atoms, blue spheres designate carbon atoms and, the small green spheres are hydrogen atoms.

that shows equol binding GPER1 in human skin. However, equol Comparative studies examining polyphenolic compounds have
has been shown to bind the orphan G-protein-coupled receptor demonstrated that equol is a superior antioxidant, having greater
(GPR30) in endothelial cells to activate endothelial NO synthase antioxidant capacity than vitamin C or vitamin E in several in vitro
(eNOS) that has a positive influence on arterial blood pressure. Thus, tests (Arora et al., 1998; Mitchell et al., 1998). In fact, in a more
equol may improve endothelial function and lower blood pressure recent study, equol exhibited one of the highest antioxidant activ-
and thus have a potential protective role in cardiovascular disease ities, when three different in vitro assays were used, and equol was
(Rowlands et al., 2011). more effective than the positive controls quercetin and ascorbic
Equol is also a selective androgen modulator (SAM), having the acid (Rufer and Kulling, 2006). Finally, equol has greater antioxidant
ability to specifically bind 5␣-dihydrotestosterone (5␣-DHT) and activity (i.e., oxidative damage to lipid membranes, etc.) compared
inhibit its potent negative actions in skin (Gopaul et al., 2012; to genistein (Mitchell et al., 1998; Rufer and Kulling, 2006).
Lephart, 2013a). Equol has the ability to bind to ERR ␥, which In other tissue sites, equol was proven to have significant
has important implications for anti-aging in human skin (Hirvonen antioxidant effects in bovine aortic endothelial cells, where equol
et al., 2011; Krahn-Bertil et al., 2008; Lephart, 2013a). Addition- protected against peroxide-induced cell death (Cheng et al., 2010).
ally, when equol is bound to ERR ␥, it decreases inflammation Additionally, equol protected against apoptosis and vascular injury
and has anti-proliferative effects on prostate and breast cancer through oxidative stress in porcine pulmonary arteries and human
cells (Hirvonen et al., 2011). Finally, equol research has dramati- pulmonary artery endothelial cells (Chung et al., 2008; Kamyama
cally increased within the past two decades, and this polyphenolic et al., 2009). Equol also significantly reduced oxidative stress and
molecule along with other botanical compounds is widely used in protected rats against focal cerebral ischemia (Ma et al., 2010).
personal care products such as skin protectants, whitening, anti- In food, a recent study examined the changes in antioxi-
wrinkle, and anti-aging ingredients as well as benefiting prostate dant compounds in white cabbage during winter storage. The
health (Baumann, 2002; Draelos and Puglises, 2011; Evans and major groups of antioxidants identified in cabbage were vitamins,
Johnson, 2010; Gopaul et al., 2012; Jackson et al., 2014; Lephart, flavonoids and phenolic acids (Hounsome et al., 2009, 2010). Of the
2013a, 2014a; Lund et al., 2011; Weber et al., 1997). isoflavonoid compounds examined, equol was identified as being
present in cabbage at high levels similar to other polyphenolic com-
pounds that can serve as an antioxidant during the storage of white
cabbage to help prevent oxidative stress (Hounsome et al., 2009,
4.1. Antioxidant properties of equol
2010).
Equol has recently caught the interest of researchers because
of its powerful antioxidant activity and its unique molecular and 4.2. Equol’s lipophilic nature and penetration into human skin via
biochemical messenger properties with implications in treating a reservoir delivery mechanism
age-related diseases (Lephart 2013a,b; Setchell and Clerici, 2010).
For example, in plants, the accumulation of polyphenolic com- The lipophilic nature of equol is shown via its octanol-water
pounds such as flavonoids and phenolic molecules have been linked partition coefficient of 3.2, which is higher than other polyphenolic
to pathogen resistance (Hounsome et al., 2009, 2010; Lephart et al., molecules (Rothwell et al., 2005). For example, the octanol-
2014). There are many polyphenolic molecules that act as antioxi- water partition coefficients for resveratrol = 3.0; genistein = 3.0 and
dants including isoflavonoids (Bohn, 2010; Lephart et al., 2014; Li diadzein = 2.5. Additionally, intestinal absorption data supports this
et al., 2012; Hounsome et al., 2009, 2010; Rice-Evans, 2001). proposition, where equol (either as the R- or S-isomer) has the high-
46 E.D. Lephart / Ageing Research Reviews 31 (2016) 36–54

Fig. 11. [3 H]-Equol percutaneous absorption profile into human skin (panel A) and Cartoon of Equol penetration into keratinocytes and pooling via a “reservoir” mechanism
in the epidermis with delayed release into the dermis over time (panel B). Panel A: [3 H]-equol was used to determine percutaneous absorption into human skin with a profile
showing an initial maximum peak flux occurring 6 h after dosing followed by a decline with a secondary lower peak at approximately 26–28 h after a single applied dose.
Human trunk skin obtained from 4 individuals (2 males and 2 females of Caucasian and Hispanic decent, ages 35–51, n = 3 per subject) were tested by Franz cell techniques
(Lephart, 2013a). The standard error of the mean (SEM) among the time point collections ranged from 0.005 to 0.024 (not shown graphically) (Lephart, 2013a). Panel B:
Cartoon display of typical active ingredients versus equol in penetrating human skin layers. Some topical dermal formulations penetrate directly through the human skin
layers (shown by dashed gray arrows), while other topical dermal formulations do not penetrate through the skin layers (shown by the blue dashed arrow). Based upon
the percutaneous absorption profile of equol through human skin it is sequestered into the epidermal compartment due to its affinity to and the abundance of estrogen
receptor beta (ER ␤) in keratinocytes (green dashed arrows) and forms an equol “reservoir” that has a time-release prolife into the dermis to act upon fibroblasts and promote
anti-aging effects in human skin.

est absorption levels (80–85%) compared with other isoflavonoids 2008; Xing et al., 2009; Zhong et al., 2009). This epidermal reser-
like genistein (at 15–20%) or daidzein (at 30–40%) (Setchell and voir delivery mechanism of equol is depicted in Fig. 11B. Finally,
Clerici, 2010; Setchell et al., 2009). Drug delivery studies have the delivery of approximately 14 nM racemic equol into the ker-
shown the more stable isomer is R-equol vs. S-equol (Alvira et al., atinocytes (after a single dose) in the percutaneous absorption
2008), and in vitro culture data in breast and prostate cancer cells studies was similar to previous in vitro culture results, where the
suggested that the R-isomer accounts for the chemo-protective continual exposure of 10 nM equol significantly stimulated col-
effects of equol rather than the S-isomer (Magee et al., 2006). lagen and elastin, while at the same time significantly inhibited
Franz cell testing using tritiated racemic equol penetration into MMPs protein expression (Gopaul et al., 2012; Lephart, 2013a).
human skin has shown: 1) the percutaneous absorption and, 2) skin
content distribution among the epidermal and dermal compart-
ments (Lephart, 2013a). As displayed in Fig. 11A, the [3 H]-equol 5. Equol’s anti-aging influence on human skin by reducing
penetrated into human skin with a profile showing an initial max- oxidative stress in cascade events
imum peak flux occurring 6 h after dosing followed by a decline
in penetration with a secondary lower peak flux at approxi- The formation of ROS is a widely accepted pivotal mecha-
mately 26–28 h after a single applied dose. Notably, when the nism leading to skin aging (Gonzaga, 2009; Kammeyer and Luiten,
actual epidermal/dermal content of the tritiated racemic equol 2015; Natarajan et al., 2014; Rittie and Fisher, 2002; Zouboulis and
was quantified, the epidermal content was higher than typically Makrantonaki, 2011). Cell culture (primary or organotypic), molec-
observed with most topical compounds or drugs representing an ular, and gene expression/array methods have examined equol’s
unusual delivery profile (Lephart, 2013a). Notably, racemic equol influence on oxidative stress along with various biomarkers at dif-
was sequestered into the epidermal compartment due, most likely, ferent cascade steps/events that lead to damaged skin. However,
to the abundance of estrogen receptor subtypes (ER ␤) in ker- since equol can be expressed as isomers, the type of equol tested
atinocytes for which S-equol has a high affinity (Gopaul et al., 2012; (racemic, S-equol or R-equol) is noted below, when this infor-
Lephart, 2013a,b; Pelletier and Ren, 2004; Setchell et al., 2005; mation is known. In a comprehensive investigation on equol as:
Thornton et al., 2003). Thus, the epidermal “reservoir” delivery R-equol, racemic equol or S-equol from PCR/mRNA studies quan-
mechanism of topically applied equol to the dermal layer over time tifying human skin gene expression, only three genes displayed
has not been seen with other compounds such other polyphenolic the greatest significant expression by S-equol, whereas, 16 genes
molecules like resveratrol along with the isoflavonoid, genistein, displayed the greatest significant levels (either stimulation or inhi-
or topical drugs like 17␤-estradiol (Chadha et al., 2011; Hung et al., bition) by R-equol and/or racemic equol, which suggest that, in
general, R-equol and/or racemic equol was more potent compared
E.D. Lephart / Ageing Research Reviews 31 (2016) 36–54 47

Fig. 12. Summary of equol’s positive influences on human skin via inhibition and/or decrease of reactive oxygen species (ROS) and/or protection against oxidative stress (OS).
Tissue Inhibitor of Matrix Metalloproteinases (TIMP), Matrix Metalloproteinases (MPPs), 5␣-dihydrotesterone (5␣-DHT), Pro-inflammatory Transcription Factor NF-kappB
(NFkappaB), Nuclear-factor-erythroid 2-related factor 2 (Nrf2), Estrogen receptor beta (ER ␤), AP-1 is a nuclear transcription element, Activator Protein − 1 (AP-1) and
Estrogen Related Receptor gamma (ERR ␥).

to S-equol for optimal human skin health (Lephart, 2013a). The protection against peroxide-induced endothelial cell apoptosis by
findings from various investigations are described below under activation of estrogen receptor and Nrf2/ARE signaling pathways.
specific headings and summarized in Fig. 12. Finally, Froyen and Steinberg (2011) reported that racemic equol
increased the expression of the xenobiotic metabolizing enzyme
5.1. Equol decreases ROS via sStimulation of Nrf2 quinone reductase (both mRNA and protein levels) via similar
molecular mechanisms involving ER ␤ and Nrf2.
Oxidative Stress via ROS production by UV-light-induced signal
cascades is known to cause skin aging and the pathology of skin
disease (Bickers and Athar, 2006; Khol et al., 2011; Naylor et al., 5.2. Equol acts as an antioxidant, stimulates
2011). Nuclear-factor-erythroid 2-related factor 2 (Nrf2) is a mas- antioxidant/detoxifying enzymes and inhibits skin aging
ter regulator of the transcriptional response to oxidative stress, biomarkers
and it is structurally and functionally conserved from insects to
humans (Lacher et al., 2015). Nrf2 plays a key role in the cellular Few in vivo studies have been performed using equol as an
defense against oxidative and xenobiotic stressors by its capac- antioxidant or protecting agent against disease. However, Ma et al.
ity to induce the expression of numerous genes, which encode (2010) examined the hypothesis that genistein and equol were
detoxifying enzymes and antioxidant proteins that provide pro- neuro-protective in transient focal cerebral ischemia in male and
tection in endothelial cells, skin morphogenesis, wound repair and ovariectomized rats by inhibiting oxidative stress. In brief, the
skin cancer (Beyer et al., 2007; Greenwald et al., 2015; Sekhar and results of this study showed that equol supplementation via the diet
Freeman, 2015; Zhang et al., 2013). Additionally, Nrf2 deficiency was more potent than genistein, in general, in preventing transient
or Nrf2-knockdown is known to cause lipid oxidation, inflamma- focal cerebral ischemia in rats by decreased brain levels of superox-
tion, and extra cellular matrix-protease expression [e.g., MMPs, ide production and NADPH oxidase (NOX) activity that are known
cyclo-oxygenases (Cox)] in UVA-irradiated skin fibroblasts or heat to increase oxidative stress. As noted by Ma et al. (2010), equol is
shock-induced human dermal fibroblasts (Gruber et al., 2015; Park a better antioxidant than genistein or daidzein, and this may assist
and Oh, 2015). in the interpretation of the obtained results as supported by other
Bottai et al. (2012) reported that 17␤-estradiol protects studies (Arora et al., 1998; Mitchell et al., 1998; Rufer and Kulling,
human keratinocytes and fibroblasts against oxidative damage 2006).
by counteracting hydrogen peroxide-mediated lipoperoxidation. In a double-blind human investigation, Pusparini and Hidayat
Furthermore, equol has been shown to increase nuclear-factor- (2015) studied the effect of soy isoflavone supplementation
erythroid 2-related factor 2 (Nrf2) (Zhang et al., 2013). (100 mg/day for 6 months) on endothelial dysfunction and oxida-
As discussed by Jackson et al. (2014), the molecular mechanism tive stress in equol-producing postmenopausal women using
involves the release of Nrf2 (transcription factor) from Keap 1, its vascular cell adhesion molecule-1 (VCAM-1) and nitric oxide (NO)
cytoplasmic binding protein and subsequent binding to the antiox- as markers of vascular endothelial function, and malonyldialde-
idant response element (ARE) that is present in the promoter region hyde (MDA) as an oxidative stress marker. The authors found
of genes for antioxidant proteins and enzymes. Specifically, equol that isoflavone supplementation in postmenopausal women with
may increase Nrf2 levels and/or bind to the estrogen-responsive equol-producer status had a beneficial effect by decreased MDA
elements (EREs) in the promoter region the Nrf2 gene and/or concentrations, but without improvement of VCAM-1 and NO
increase gene expression of other antioxidant genes. In support concentrations. Thus, the results of this study provided potential
of this concept, Zhang et al. (2013) showed that S-equol provided information that decreased oxidative stress exists in equol-
48 E.D. Lephart / Ageing Research Reviews 31 (2016) 36–54

producing postmenopausal women with dietary soy isoflavone stimulate the gene expression of PCNA that is known for its protec-
supplementation. tive skin effects (Gopaul et al., 2012; Lephart, 2013a).
One small human in vitro study reported that R-equol pro- Several reports have shown that nerve growth factor (NGF) is
tected gastric cells from oxidative stress and induced cell death important in cell survival, wound healing by stimulation of collagen
(Asciutti et al., 2010). Another in vitro study reported that racemic and regeneration of cutaneous nerves (Marconi et al., 2003; Nithy
equol inhibited LPS-induced oxidative stress and at the same time et al., 2003; Yaar et al., 1991; Zhai et al., 1996). It is known that
enhanced the immune response in chicken macrophages (Gou et al., NGF expressed in keratinocytes is reduced by UVB exposure (by
2015). sun/photo-aging) (Marconi et al., 2003). Gopaul et al. (2012) and
ROS generation sits at the top of the pathway of the oxidative Lephart (2013a) showed that racemic equol stimulated NGF gene
stress cascade, and oxidative stress can be considered the com- expression in human skin that suggested protective dermal effects
mon denominator of both intrinsic and extrinsic aging by oxidative by NGF from oxidative stress.
metabolism and exposure to UVR, respectively. Previous studies
have examined the role of antioxidants in dermal aging by ROS 5.4. Equol inhibits AP-1 and neoplastic cell growth
production via gene expression analysis (Avantaggiato et al., 2014;
Pandel et al., 2013). As pointed out above, equol is a strong antiox- AP-1 is a nuclear transcription element that is part of the oxida-
idant itself. Racemic equol has been shown to stimulate the gene tive stress cascade. It induces MMPs in human skin that in turn
expression of antioxidant enzymes in human skin such as SOD 2 and activates collagen degradation (Huang et al., 1997). AP-1 also blocks
thioredoxin reductase 1 (TXNRD 1) (Gopaul et al., 2012; Lephart, the positive pro-collagen actions of transforming growth factor-␤1
2013a,b). SOD 2 and TXNRD 1 are known to protect against free red- (Fisher et al., 1996). Kang et al. (2007a) reported that the anti-tumor
icals, oxidiative stress and UV-induced skin damage (Shindo et al., effects of racemic equol are due to the inhibition of cell transfor-
1994; Velarde et al., 2012). In addition to dermal studies, equol had mation by the MEK signaling pathway by blocking AP-1. Racemic
been shown to inhibit peroxide-induced bovine aortic endothelial equol dose-dependently attenuated TPA-induced activation of AP-
cell death demonstrating equol has significant antioxidant effects 1, whereas daidzein did not exert any effect when tested at the same
to neutralize hydrogen peroxide (Chung et al., 2008). Furthermore, concentrations (Kang et al., 2007a). Finally, ER ␤ signaling has been
Widyarini et al. (2012) reported the positive biological activities shown to protect against transplanted skin tumor growth in mice
of equol that include the UV-protective antioxidant effects via the (Cho et al., 2010), which implicated a common mechanism of how
endogenous cutaneous antioxidant enzyme haem oxygenase (HO)- the blocked AP-1 actions by equol reported by Kang et al. (2007a)
1. Haem oxygenase is a protector of lipid peroxidation and is part of may be mediated.
the mitogen activated protein kinase (MAPK) pathway that results Additionally, equol has been shown to bind to ERR ␥ that in turn
in the activation of antioxidant genes/enzyme via the Nrf2/KEAP enhanced the transcriptional activity of ERR ␥, which is known to
1/ARE pathway (Mann et al., 2009). Also, racemic equol stimu- protect against neoplastic growth (Hirvonen et al., 2011). Equol’s
lated the gene expression of the SH-rich detoxifying proteins like precursor molecule daidzein did not have this effect (Krahn-Bertil
metallothionein-1H and metallothionein-2H that protect against et al., 2008). It is also known that ERR ␥ is present in keratinocytes
metal toxicity (Gopaul et al., 2012; Lephart, 2013a). Previous stud- and fibroblasts in human skin, where its actions may show sim-
ies have shown that equol’s photo-protective effect in mouse and ilar favorable protection as that seen against breast and prostate
human skin is dependent on metallothionein (Widyarini et al., cancers (Hirvonen et al., 2011; Krahn-Bertil et al., 2008).
2006b). Moreover, racemic equol was shown to inhibit the gene Finally, it has been demonstrated that ER ␤ signaling in the
expression of human skin aging biomarkers: S100 calcium-binding prostate (and breast tissue) are associated with decreased inflam-
protein (CBP) A8 and CBP A9 that are known to increase with skin mation and neo-plastic growth (Lephart, 2014a). S-Equol is known
aging (Gopaul et al., 2012). to have a high affinity for ER ␤, which is the predominate ER in ker-
Also, racemic equol was reported to inhibit the gene expres- atinocytes and fibroblasts in human skin (Pelletier and Ren, 2004;
sion of type 1 5␣-reductase in human skin (Gopaul et al., 2012; Setchell et al., 2005; Thornton et al., 2003) and this ER signaling
Lephart, 2013a). Recall, this enzyme converts testosterone to the mechanism protects against immune suppression by UV radia-
more potent androgen, 5␣-DHT in dermal fibroblasts that cause the tion exposure (Chang et al., 2010; Pomari et al., 2015; Widyarini
stimulation of MMPs and reduces tissue wound repair (Ma et al., et al., 2006a). The high affinity for ER ␤ by S-equol may account for
2010; Makrantonaki and Zouboulis, 2009; Nitsch et al., 2004). In the unique topical dermal absorptive and ‘reservoir’ penetration
cell cultures of human dermal fibroblasts 5␣-DHT was cytotoxic method into human skin that may account, in part, for its positive
at 10 nM (Gopaul et al., 2012). In this regard, clinical evidence has benefits (Lephart, 2013a).
demonstrated that the accumulation of 5␣-DHT in dermal papilla
cells is implicated in androgenetic alopecia via the induction of ROS 5.5. Equol inhibits NFkappaB
leading to cell senescence and cell death (Lee et al., 2015).
Previous studies have demonstrated racemic equol’s ability to The pro-inflammatory transcription factor NF-kappB has been
specifically bind to 5␣-DHT and prevent its negative biological studied for more than 30 years. It was first discovered by Sen and
actions such as skin aging (Gopaul et al., 2012; Makrantonaki and Baltimore in 1986 (Gupta et al., 2010). NF-kappaB remains an excit-
Zouboulis, 2009; Nitsch et al., 2004). Finally, equol’s positive effects ing and active area of investigation, where it is expressed in all cell
not only have applications to human skin, but prostate health as types and is evolutionarily conserved (Ghosh et al., 1998). NFkap-
well (Lephart, 2014a; Lund et al., 2011). paB is involved in the oxidative stress mechanism by the expression
of numerous genes such as the cytokines and plays a major role in
5.3. Equol protects DNA and enhances nerve and tissue repair the pathology of inflammatory disease (Gilmore, 2006; Ghosh et al.,
1998; Hayden and Ghosh, 2012; Schreck et al., 1991).
ROS production is known to damage DNA, proteins and lipids Several investigators examined the inhibition of NFkappB by
and other cellular components (Bickers and Athar, 2006; Gonzaga, equol. Kang et al. in (2005) showed that racemic equol inhibited
2009; Khol et al., 2011). In human skin an important aging tumor necrosis factor-␣ gene expression by blocking NFkappB in
biomarker is proliferating cell nuclear antigen (PCNA). It is involved mouse macrophages that was independent of an estrogen recep-
in DNA repair, and it is known to decrease in aged skin (Goukassian tor mechanism. Tumor necrosis factor-␣ is a cell signaling protein
et al., 2000; Takahashi et al., 2005). Racemic equol was shown to (cytokine) involved in systemic inflammation (Kang et al., 2005).
E.D. Lephart / Ageing Research Reviews 31 (2016) 36–54 49

Furthermore, Kang et al. in (2007b) reported the dose-dependent MMPs and elastase has profound effects on maintaining the major
inhibitory effects of racemic equol (by in vivo administration) on extra cellular proteins (i.e., collagen and elastin) in opposition to
nitric oxide (NO) production and inducible nitric oxide synthase the insult of oxidative stress.
(iNOS) gene expression in murine macrophages from lipopolysac-
charide (LPS)-treated mice. The enzyme NO synthase (NOS) 5.8. Equol stimulates TIMP 1
generates NO that is a free radical, and the overproduction of NO
by iNOS is associated with the development of several diseases Since oxidative stress directly or indirectly activates MMPs, the
including atherosclerosis, stroke, septic shock and Alzheimer’s dis- tissue inhibitors of matrix metalloproteases (TIMPs) are impor-
ease (Kang et al., 2007b). The gene expression of iNOS is regulated tant skin enzymes that inhibit the actions of MMPs (Draelos and
mainly at the transcriptional level in macrophages and NF-kappB Puglises, 2011; Fazio et al., 1988; Freedberg et al., 2003). The
via the MAPK pathway and PI3K/Akt pathways. The MAPK pathway TIMPs are known to decrease with exposure to UV light and with
has been well known to be involved in the regulation of iNOS gene aging that contributes to dermal matrix degradation, impaired cell
expression and NF-kappaB activation (Kang et al., 2007b). In this growth and survival (Farage et al., 2010; Hornebeck, 2003). It is
study, LPS-induced activation of Akt was suppressed by racemic known that increased TIMP expression and activation blocks the
equol, and it also blocked LPS-induced NFkappaB activation. negative influence of MMPs in causing skin damage. When the iso-
mers of equol along with racemic equol were examined in gene
5.6. Equol inhibits the inflammatory response arrays using human epidermal/dermal skin equivalents, racemic
equol showed the highest stimulation of TIMP 1 followed by S-
Extensive research during the last two decades has revealed the equol, whereas, R-equol did not stimulate TIMP 1 at all (Lephart,
mechanism by which continued oxidative stress leads to chronic 2013a). This characterization of the equol isomer’s impact on TIMP
inflammation, which in turn, mediates most chronic diseases 1 demonstrated the complex manner of extracellular dermal matrix
including cancer and skin damage (Bar-Or et al., 2015; Bickers and regulation of collagen protection and maintenance.
Athar, 2006; Droge, 2002; Maes et al., 2011; Maritim et al., 2003;
Valko et al., 2007). Recent research on polyphenolic molecules from
5.9. Equol stimulates collagen
plants sources has expanded the horizon for potential therapeutic
remedies and treatments (Evans and Johnson, 2010; Adlercreutz
Collagen is the most important extracellular dermal matrix pro-
et al., 2004; Park and Pezzuto, 2015). It is well documented that var-
tein that maintains optimal dermal health (Draelos and Puglises,
ious pro-inflammatory markers in human skin are increased with
2011; Farage et al., 2010). While oxidative stress via a cascade
UV exposure (Bickers and Athar, 2006; Khol et al., 2011; Strickland
mechanism degrades collagen and reduces the deposition of col-
et al., 1997).
lagen (via MMPs) in the dermal layer, it is important not only to
It has been shown that racemic equol inhibited the gene expres-
block the negative insults of ROS production, but to enhance colla-
sion of several pro-inflammatory biomarkers such as interleukin-1
gen deposition. In both cell culture and gene array studies, racemic
alpha (IL-1A), IL-6, IL-8 and interleukin-1 receptor 2 as well as COX-
equol stimulated pro-collagen type 1 and collagen levels (Gopaul
1 and tumor necrosis factor receptor (Gopaul et al., 2012; Lephart,
et al., 2012; Lephart, 2013a). When the equol isomers were exam-
2013a). These pro-inflammatory biomarkers are known to increase
ined in human skin gene expression experiments, racemic equol
with UV exposure and aging (Bickers and Athar, 2006; Natarajan
stimulation of collagen was highest followed by R-equol and both
et al., 2014; Strickland et al., 1997).
were significantly higher than S-equol (Lephart, 2013a).
It is also known that ER ␤ signaling protects epidermal cytokine
expression and immune function by UVB exposure (Chang et al.,
2010; Cho et al., 2008; Widyarini et al., 2012; Widyarini et al., 5.10. Equol stimulates elastin
2006a). Since racemic equol has been utilized in most research
investigations, the S-equol in racemic equol may act by bind- Just as the case with collagen, oxidative stress via a cascade
ing to ER ␤ receptors in keratinocytes. This may explain the mechanism degrades elastin and reduces the deposition of elastin
obtained gene expression results, where equol inhibited several (via MMPs, fibroblast collapse/elastase) in the dermal layer, it is
pro-inflammatory biomarkers (Gopaul et al., 2012). However, a important not only to block the negative insults of ROS produc-
comprehensive study that examined the equol isomers along with tion, but to enhance elastin deposition (Draelos and Puglises, 2011;
racemic equol in human skin via gene array analysis suggested Farage et al., 2008; Mera et al., 1987; Tulah and Birch-Machin,
that R-equol and/or racemic equol are better inhibitors of the 2013). While collagen fibers provide the structural framework of
pro-inflammatory biomarkers (Lephart, 2013a). This suggests that the skin, elastin provides the “bounce back” or the elasticity of the
further research is warranted to determine the mechanism of how skin that prevents the rapid increase in wrinkle formation and sag-
equol (isomers) inhibit pro-inflammatory markers. ging that is a hallmark of skin aging and dermal damage. In both
cell culture and gene array studies, racemic equol stimulated elastin
5.7. Equol inhibits MMPs and elastase levels (Gopaul et al., 2012). Surprisingly, racemic equol stimulated
elastin to a higher level compared to the endogenous steroid hor-
It is known that oxidative stress directly and/or indirectly stim- mone, 17␤-estradiol (Lephart, 2013a). In the examination of the
ulates the production of MMPs and elastase, that in turn breakdown equol isomers in human skin gene expression experiments racemic
collagen and elastin in the dermal matrix (Bickers and Athar, 2006; equol displayed the greatest stimulation of elastin followed by R-
Farage et al., 2008, 2010; Mera et al., 1987; Tulah and Birch-Machin, equol, whereas, S-equol did not stimulate elastin at all (Lephart,
2013). Racemic equol was examined in two different studies for 2013a). A summary of equol’s positive influences on human skin in
its influence on human skin gene expression of these important reference to ROS and oxidative stress effects is shown in Fig. 12.
biomarkers. Racemic equol inhibited the gene expression of MMP 1,
MMP 3, and MMP 9 (Gopaul et al., 2012; Lephart, 2013a). In 8-week 6. Equol: comparison to resveratrol
organotypic cell cultures of human dermal fibroblasts, racemic
equol decreased the expression of the elastase enzyme by 35%, When the above equol skin biomarker results are compared to
while the potent natural steroid hormone, 17␤-estradiol decreased the botanical that has the highest recognition by the general pub-
the expression by 8.4% (Gopaul et al., 2012). Equol’s ability to inhibit lic or lay profile namely, resveratrol (Lephart et al., 2014; Park
50 E.D. Lephart / Ageing Research Reviews 31 (2016) 36–54

Table 1
Comparison of the chemical properties and actions of equol to resveratrol in human skin.

Equol Resveratrol

1. Molecular Weight 242.2 g/mol 228.2 g/mol


2. Octanol-water partition CLogP 3.2 CLogP 3.0
3. Estrogen Receptor (ER) Binding ER ␤ > ER ␣ mixed ER agonist
4. Directly Binds 5␣-DHT Yes No
5. Inhibits 5␣-Reductase Enzyme Yes No
6. Activates Estrogen-Related Receptor Gamma (ERR␥) Yes No
7. Topical Reservoir Delivery Yes No
8. Stimulates ECM proteins at [nM] Yes No at [␮M]
9. Directly Stimulates Sirutins No Yes
10. Dietary Supplementation-Decreases Facial Wrinkles Yes Unknown

CLogP = hydrophilicity index; 5␣-DHT = 5␣-dihydrotestosterone; [nM] = nanomolar concentration; [␮M] = micromolar concentration.

and Pezzuto, 2015), equol displayed better biochemical, molec- S-equol reduced facial wrinkles compared to placebo controls with-
ular, protective and delivery mechanisms of action to promote out adverse hormonal or gynecological effects. While there is ample
and/or improve human skin health as a topical anti-aging ingredi- evidence that equol has the potential to decrease oxidative stress,
ent/compound (Lephart, 2013a). This is interesting since equol and further research is required to determine the exact mechanisms of
resveratrol have similar molecular weights and octanol-water par- how equol protects human skin from the damaging effects of ROS.
titions (indicating lipid solubility). However, equol directly binds Finally, it is interesting to consider how polyphenolic molecules via
the potent androgen 5␣-DHT, inhibits the 5␣-reductase enzyme dietary supplementation or functional foods may prevent oxida-
in fibroblasts, acts as a selective SERM modulator by bind ER␤ tive stress to ameliorate cellular dermal damage (Lobo et al., 2010;
with higher affinity compared to ER␣ and binds and activates Poljsak and Dehmane, 2012).
estrogen related-receptor gamma, while resveratrol does not have
these properties. This may account for equol’s ability to stimulate 7. Summary and conclusions
collagen in human dermal fibroblast cell cultures at nano-molar
concentrations, whereas, resveratrol requires micro-molar levels Oxygen in biology is essential for life, but it comes at a cost,
to increase collagen. Also, equol is highly absorbed after oral dos- where ROS are generated by oxidative metabolism via normal cellu-
ing (>80%), whereas resveratrol is rapidly metabolized which make lar function. Human skin exposed to solar UV radiation dramatically
this route of delivery challenging from a commercial perspective. In increases ROS production and oxidative stress. It is important to
fact, dietary supplementation of equol appears to provide an effec- understand the characteristics of human skin and how dermal
tive route of delivery in the clinical application of decreasing facial damage occur with chronological (intrinsic) aging and photo-aging
wrinkles (see below). See Table 1 for a descriptive comparison of (extrinsic aging) via the impact of ROS production and oxidative
the chemical properties and actions of equol to that of resveratrol stress mechanisms. This includes a cascade of events that involve
in human skin. a variety of cell/molecular signaling pathways. However, it is well
In this regard, resveratrol is a known sirtuin 1 (SIRT1) or anti- established that photo-aging accounts for a majority of the patho-
aging activator, whereas, equol is not. The sirtuins are a class of logical changes in human skin and is dependent upon the exposure
NAD+ -dependent protein deacetylase enzymes that regulate a wide to and intensity of solar UV radiation. The oxidative stress effects
variety of cellular activities, which promote cell survival and delay on skin aging involve damage to DNA, the inflammatory response,
or attenuate many age-related changes such as preventing early reduced production of antioxidants and activation/inhibition of
mortality in obese animals (Lephart et al., 2014; Park and Pezzuto, various signaling factors that ultimately lead to the production
2015). Thus, activators of sirtuins may benefit fundamental cellu- of matrix metalloproteinases (MMPs) that degrade collagen and
lar processes that protect cells from stress and potentially treat elastin in the dermal skin layer. The outcome of this complex aging
age-related conditions and lengthen a healthy life (Lephart et al., process is the disruption of the epidermal/dermal matrix, which
2014) (see Table 1). In a recent study, the combined administration has the highest antioxidant content and immune defense function
of equol with resveratrol enhanced SIRT1 expression in endothe- in the body. The visually perceived appearance of wrinkles, age-
lial cells demonstrating equol’s unique beneficial biological actions spots and loss of skin tone reflect the aging process. The goal is to
(Davinelli et al., 2013). delay the onset of aging and/or slow down skin aging with time and
Equol is currently used in skin treatments, and the positive influ- maintain good dermal health.
ences on gene and protein expression (anti-aging, anti-oxidant and Botanicals, as active ingredients, represent one of the largest
anti-inflammatory properties) have been reported (Gopaul et al., percentages of compounds in use in dermatology/cosmeceuticals
2012). Recently, it has been proposed that ER ␤ agonists like S-equol today to combat aging. An emerging botanical is equol. Equol is a
may have better beneficial dermal applications (Chang et al., 2010), polyphenolic, isoflavonoid molecule found in plants/food products
compared to R-equol or racemic equol, however, this proposition and is a metabolite of daidzein by intestinal bacteria in all humans
has not been confirmed by the available human skin gene expres- and animals. Equol has the potential to decrease oxidative stress
sion data (Gopaul et al., 2012; Lephart, 2013a). A report by Oyama and increase skin cellular longevity in human skin. The characteris-
et al. (2012) suggested that isoflavone dietary supplementation tics of equol and evidence of its protective properties and biological
(for 12 weeks) in postmenopausal Japanese women that contained actions are discussed in reference to ROS generation and oxidative
E.D. Lephart / Ageing Research Reviews 31 (2016) 36–54 51

stress events. Some of these topics include equol’s action of: stimu- Becker, B.F., 1993. Towards the physiological function of uric acid. Free Radic. Biol.
lation of Nrf2, antioxidant/detoxifying enzymes, and extra cellular Med. 14, 615–631.
Beyer, T.A., Keller, U., Braun, S., Schafer, M., Werner, S., 2007. Roles and
matrix proteins along with DNA and tissue repair and, equol’s mechanisms of action of the Nrf2 transcription factor in skin morphogenesis:
inhibition of: NFkappaB, pro-inflammatory biomarkers and MMPs. wound repair and skin cancer. Cell Death Differ. 14, 1250–1554.
Many phyto-chemical effects on the skin are still unknown, and Bickers, D.R., Athar, M., 2006. Oxidative stress in the pathogenesis of skin disease. J.
Invest. Dermatol. 126, 2565–2575, http://dx.doi.org/10.1038/sj.jid.5700340.
further research is warranted to clarify how they protect against Bohn, T., 2010. Isoflavone bioavailability from foods and supplements dietary
oxidative stress in skin aging and damage not only via topical appli- factors impacting utilization. Argo Food Ind. Hi-Technol. 21, 59–62.
cations but by dietary supplementation and/or functional foods. Borras, C., Gambini, J., Gomez-Cabrera, M.C., Sastre, J., Pallardo, F.V., Mann, G.E.,
Vina, J., 2005. 17beta-oestradiol up-regulates longevity-related, antioxidant
enzyme expression via the ERK1 and ERK2[MAPK]/NFkappaB cascade. Aging
Conflict of interests Cell 4, 113–118.
Bottai, G., Mancina, R., Muratori, M., Di Gennaro, P., Lotti, T., 2012. 17␤-estradiol
protects human skin fibroblasts and keratinocytes against oxidative damage. J.
The author declares no conflict of interest in the data/research Eur. Acad. Dermatol. Venereol. 27, 1236–1243,
presented in this review and regarding the publication of this 10.111/j.1468-3083.2012.0469.x.
Brincat, M.P., Baron, Y.M., Galea, R., 2005. Estrogens and the skin. Climacteric 8,
manuscript. The author has equol patents (U.S. and worldwide). 110–113.
Buonocore, G., Perrone, S., Tataranno, M.L., 2010. Oxygen toxicity: chemistry and
biology of reaction oxygen species. Semin. Fetal Neonatal Biol. 15, 186–190.
Acknowledgement Burns, D.A., Breathnach, S.M., 2004. Rook’s Textbook of Dermatology, 1, 3.1 and
4.1., 7th ed. Blackwell Publishing Co., New York.
Burton, G.J., Jauniaux, E., 2011. Oxidative stress. Best Pract. Res. Clin. Obstet.
This study/review was supported, in part, by LS/TTO funding,
Gynaecol. 25, 287–299, http://dx.doi.org/10.1016/j.bpobgyn.2010.10.016.
19-2215 at Brigham Young University. Chadha, G., Sathigari, S., Parsons, D.L., Jayachandra, B.R., 2011. In vitro
percutaneous absorption of genistein from topical gels through human skin.
Drug Dev. Ind. Pharm. 37, 498–505.
References Chang, K.C., Wang, Y., Oh, I.G., Freedman, L.P., Thompson, C.C., Chung, J.H., Nagpal,
S., 2010. Estrogen receptor beta is a novel therapeutic target for photoaging.
Abiru, Y., Kumenura, M., Ueno, T., Uchiyama, S., Masaki, K., 2012. Discovery of an Mol. Pharm. 77, 744–750.
S-equol rich food stinky tofu, a traditional fermented soy product in Taiwan. Cheng, C., Wang, X., Weakley, S.M., Kougias, P., Lin, P.H., Yao, Q., Chen, C., 2010. The
Int. J. Food Sci. Nutr. 63, 964–970. soybean isoflavonoid equol blocks Ritonavir-induced endothelial dysfunction
Adlercreutz, H., Heinonen, S.M., Penalvo-Garcia, J., 2004. Phytoestrogens, cancer in porcine pulmonary arties and human pulmonary artery endothelial cells. J.
and coronary heart disease. Biofactors 22, 229–236. Nutr. 140, 12–17, http://dx.doi.org/10.3945/jn.109.110981.
Akazaki, S., Nakagawa, H., Kazama, H., 2002. Age-related changes in skin wrinkles Chignalia, A.Z., Schuldt, E.Z., Camargo, L.L., Montezano, A.C., Callera, G.E., Laurindo,
assessed by a novel three-dimensional morphometric analysis. Br. J. Dermatol. F.R., Lopes, L.R., Avellar, M.C.W., Carvalho, M.H.C., Fortes, Z.B., Touyz, R.M.,
147, 689–695. Tostes, R.C., 2012. Testosterone induces vascular smooth muscle cell migration
Akhtar, K., Broekelmann, T.J., Miao, M., Keeley, F.W., Starcher, B.C., Pierce, R.A., by NADPH oxidase and c-Src–Dependent pathways. Hypertension 59,
Mecham, R.P., Adair-Kirk, T.L., 2010. Oxidative and nitrosative modifications of 1263–1271.
tropoelastin prevent elastic fiber assembly in vitro. J. Biol. Chem. 285, Cho, J.-L., Allanson, M., Domanski, D., Arun, S.J., Reeve, V.E., 2008. Estrogen
37396–37404. receptor-beta signaling protects epidermal cytokine expression and immune
Alvira, E., Mayoral, J.A., Garica, J.I., 2008. Enantiodiscrimination of equol in function for UVB-induced impairment in mice. Photochem. Photobiol. Sci. 7,
␤-cyclodextrin: an experimental and computational study. J. Inclusion 120–125.
Phenom. 60, 103–113, http://dx.doi.org/10.1007/s10847-007-9358-4. Cho, J.-L., Allanson, M., Domanski, D., Arun, S.J., Reeve, V.E., 2010. Oestrogen
Ames, B.N., Cathcart, R., Schwiers, E., Hochstein, P., 1981. Uric acid provides an receptor-beta signaling protects against transplanted skin tumor growth in the
antioxidant defense in humans against oxidant- and radical-caused aging and mouse. Photochem. Photobiol. Sci. 9, 608–614.
cancer: a hypothesis. Proc. Natl. Acad. Sci. U. S. A. 78, 6858–6862. Chung, J.E., Kim, S.Y., Jo, H.H., Hwang, S.J., Chae, B., Kwon, D.J., Lew, Y.O., Lim, Y.T.,
Anderson, A., Bowman, A., Boulton, S.J., Manning, P., 2014. A role for human Kim, J.H., Kim, E.J., Kim, M.R., 2008. Antioxidant effects of equol on bovine
mitochondrial complex II in the production of reactive oxygen species in aortic endothelial cells. Biochem. Biophy. Res. Commun. 375, 420–424, http://
human skin. Redox Biol. 2, 1016–1022, http://dx.doi.org/10.1016/j.redox.2014. dx.doi.org/10.1016/j.bbrc.2008.08.027.
08.005. Common, R.H., Ainsworth, L., 1961. Identification of equol in the urine of the
Anderson, R.R., 2012. Cosmegizmoceuticals: the physics and chemistry of looking domestic fowl. Biochim. Biophys. Acta 53, 403–404, http://dx.doi.org/10.1016/
better. December 6, NY, NY In: Society of Cosmetic Chemists Annual Meeting, 0006-3002(61)90452-8.
43. Davinelli, S., Nadia, S., Visentin, M., Zella, D., Scapagnini, G., 2013. Enhancement of
Andres, A., Cleves, M.A., Bellando, J.B., Pivik, R.T., Casey, P.H., Badger, T.M., 2012. mitochondrial biogenesis with polyphenols: combined effects of resveratrol
Developmental status of 1-year-old infants fed breast milk, cow’s milk and equol in human endothelial cells. Immun. Aging 10, 29, http://dx.doi.org/
formula, or soy formula. Pediatrics 129, 1134–1140, http://dx.doi.org/10.1542/ 10.1186/1742-4933-10-28.
peds. 2011–3121. Degen, G.H., Balaszkewicz, M., Shi, L.J., Buyken, A.E., Remer, T., 2011. Urinary
Archer, D.F., 2012. Postmenopausal skin and estrogen. Gynecol. Endocrinol. 28, 2–6. isoflavone phytoestrogens in German children and adolescents- a longitudinal
Arora, A., Nair, M.G., Strasburg, G.M., 1998. Antioxidant activities of isoflavones and examination in the DONALD cohort. Mol. Nutr. Food Res. 55, 359–367, http://
their biological metabolites in a liposomal system. Arch. Biochem. Biophys. dx.doi.org/10.1002/mnfr.201000325.
356, 133–141. Djuric, Z., Chen, G., Doerge, D.R., Heilburn, L.K., Kucuk, O., 2001. Effect of soy
Asciutti, S., Setchell, K.D.R., Gizzi, S., Clerici, C., 2010. S1689 R-Equol protects isoflavone supplements on markers of oxidative stress in men and women.
human gastric cells from oxidative stress-induced cell death. Gastroenterology Cancer Lett. 172, 1–6.
138 (Suppl. 1), S-254, 10/1016/S0016-5085(10)61159-6. Draelos, Z.D., Puglises, P.T., 2011. Physiology of the Skin, 3rd ed. Allurebooks, Carol
Ashworth, J.L., Murphy, G., Rock, M.J., Sherratt, M.J., Shapiro, S.D., Shuttleworth, Stream, IL, USA.
C.A., Kieltry, C.M., 1999. Fibrillin degradation by matrix metalloproteinases: Droge, W., 2002. Free radicals in the physiologic control of cell function. Physiol.
implications for connective tissue remodeling. Biochem. J. 340, 171–181. Rev. 82, 47–95.
Avantaggiato, A., Bertuzzi, G., Vitiello, U., Iannucci, G., Pasin, M., Cervelli, V., Carinci, Duncan, K.O., Leffell, D.J., 1997. Preoperative assessment of the elderly patient.
F., 2014. Role of antioxidants in dermal aging: an in vitro study by q-RT-PCR. Dermatol. Clin. 15, 583–593.
Aesthet. Plastic Surg. 38, 1011–1016, http://dx.doi.org/10.1007/s00266-014- Escoffier, C., de Rigal, J., Rachdfore, A., Vasselet, R., Lefeque, J.L., Agache, P.G., 1989.
0380-9. Age-related mechanical properties of human skin: an in vivo study. J. Invest.
Badger, T.M., Gilchrist, J.M., Pivik, R.T., Andres, A., Shandar, K., Chen, J.-R., Ronis, Dermatol. 93, 353–357.
M.J., 2009. The health implications of soy infant formula. Am. J. Nutr. 89, Evans, J.A., Johnson, E.J., 2010. The role of phytonutrients in skin health. Nutrients
1668S–1672S, http://dx.doi.org/10.3945/ajcn.2009.26736U. 2, 903–928.
Bannwart, C., Adlercreutz, H., Fotsis, T., Wähälä, K., Hare, T., Barrow, G., 1986. Farage, M.A., Miller, K.W., Elsener, P., Maibach, H.I., 2008. Intrinsic and extrinsic
Identification of isoflavonic phytoestrogens and lignans in urine of human and factors in skin aging: a review. In. J. Cosmet. Sci. 30, 87–95.
in cow milk by GC/MS. Adv. Mass Spec. 10, 622–661. Farage, M.A., Miller, K.W., Maibach, H.I., 2010. Degenerative changes in aging skin.
Bansal, N., Parle, M., 2010. Soybean supplementation helps reverse age- and In: Farage, M.A., Miller, K.W., Maibach, H.I. (Eds.), Textbook of Aging Skin.
scopolamine-induced memory deficits in mice. J. Med. Food 13, 1293–1300, Springer-Verlag, Berlin Heidelberg, pp. 225–235.
http://dx.doi.org/10.1089/jmf.2010.1132. Fazio, M.J., Olsen, D.R., Kuizaniemi, H., Chu, M.L., Davidson, J.M., Rosenbloom, J.,
Bar-Or, D., Bar-Or, R., Rael, L.T., Brody, E.N., 2015. Oxidative stress in severe acute Uitto, J., 1988. Isolation and characterization of human elastin cDNAs: and
illness. Redox Biol. 4, 340–345, 10/1016/j/redox.2015.01.006. age-associated variation in elastin gene expression in cultured skin fibroblasts.
Baumann, L., 2002. Cosmetic Dermatology: Principles and Practice. McGraw Hill, Lab. Invest. 58, 270–277.
Hong Kong.
52 E.D. Lephart / Ageing Research Reviews 31 (2016) 36–54

Fisher, G.J., Datta, S.C., Taylor, H.S., 1996. Molecular basis of sun induced premature Huang, C., Schmid, P.C., Ma, W.Y., Schmidt, H.H., Dong, Z., 1997.
skin aging and retionoid antagonism. Nature 379, 335–339. Phosphatidylinositol-3 kinase is necessary for 12-O-tetradecanoylphorbol
Frankenfeld, C.L., 2011. Dairy consumption is a significant correlate of urinary 13-acetate-induced cell transformation and activated protein 1 activation. J.
equol concentration in representative sample of US adults. Am. J. Clin. Nutr. 93, Biol. Chem. 272, 4187–4194.
1109–1116, http://dx.doi.org/10.3945/ajcn.111.011825. Hung, C.-F., Lin, Y.-K., Huang, Z.-R., Fang, J.Y., 2008. Delivery of resveratrol, a red
Freedberg, I.M., Eisen, A.Z., Wolff, K., Austen, K.E., Goldsmith, L.A., Katz, S.I., 2003. wine polyphenol, for solutions and hydrogels via the skin. Biol. Pharm. Bull. 31,
Fritzatrick’s Dermatology in General Medicine, 6th ed. McGraw Hill, New York. 955–962.
Friedman, O., 2005. Changes associated with the aging face. Facial Plast. Surg. Clin. Inoue, T., Miki, Y., Abe, K., Haton, M., Hosaka, M., Kariya, Y., Kakuo, S., Fujimura, T.,
North Am. 13, 371–380. Hachiya, A., Aiba, S., Sasano, H., 2011. The role of estrogen-metabolizing
Froyen, E.B., Steinberg, F.M., 2011. Soy isoflavones increase quinone reductase in enzymes and estrogen receptors in human epidermis. Mol. Cell. Endocrinol.
heap-1c1c7 cells via estrogen receptor beta and nuclear factor erythroid 344, 35–40, http://dx.doi.org/10.1016/j.mce.2011.06.015.
2-related factor 2 binding to the antioxidant response element. J. Nutr. Jackson, R.L., Greiwe, G.S., Schwen, R.J., 2011. Ageing skin: oestrogen receptor ␤
Biochem. 22, 843–848. agonists offer an approach to change the outcome. Exp. Dermatol. 20, 879–882.
Fujimura, T., Haketa, K., Hotta, M., Kitahara, T., 2007. Loss of skin elasticity Jackson, R.L., Greiwe, J.S., Schwen, R.J., 2014. S-equol, an antioxidant metabolite of
precedes to rapid increase of wrinkle levels. J. Dermatol. Sci. 47, 233–239. soy dadizein, and oxidative stress in aging: a focus on skin and the
Ghosh, S., May, M.J., Kopp, E.B., 1998. NF-kB and Rel Proteins: evolutionarily cardiovascular system. In: Aging: Oxidative Stress and Dietary Antioxidants.
conserved mediators of immune responses. Ann. Rev. Immunol. 16, 225–260, Elsevier Inc., London, UK, pp. 145–155.
http://dx.doi.org/10.1146/annurev.immunol.16.1.225. Jain, T., Klein, N.A., Lee, D.M., Sluss, P.M., Soules, N.R., 2003. Endocrine assessment
Gilchrest, B.A., 1989. Skin aging and photoaging: an overview. J. Am. Acad. of relative reproductive age in normal eumenorrheic younger and older
Dermatol. 21, 610–613. women across multiple cycles. Am. J. Obstet. Gynecol. 189, 1080–1084.
Gilchrest, B.A., 2008. Sun exposure and vitamin D sufficiency. Am. J. Clin. Nutr. 88, Jou, H.-J., Tsai, P.-J., Tu, J.-H., Wu, W.-H., 2013. Stinky tofu as a rich source of
570S–577S. biavailable S-equol in Asian diets. J. Funct. Foods 5, 651–659.
Gilchrist, J.M., Moore, M.B., Andres, A., Estroff, J.A., Badger, T.M., 2010. Kammeyer, A., Luiten, R.M., 2015. Oxidative events and skin aging. Ageing Res. Rev.
Ultrasonographic patterns of reproductive organs in infants fed soy formula: 21, 16–29, http://dx.doi.org/10.1016/j.arr.2015.01.001.
comparisons to infants fed breast milk and milk formula. J. Pediatr. 156, Kamyama, M., Kishimoto, Y., Tani, M., Utsunomiya, K., Kondo, K., 2009. Effects of
215–220, http://dx.doi.org/10.1016/j.jpeds.2009.08.043. equol on oxidized low-density lipoprotein-induced apoptosis in endothelial
Gilmore, T.D., 2006. Introduction to NF-kB: players, pathways, perspectives. cells. J. Atheroscler. Thromb. 16, 239–249.
Oncogene 25, 6680–6684, http://dx.doi.org/10.1038/sj.onc.1209954. Kang, J.S., Yoon, Y.D., Han, M.H., Han, S.B., Lee, K., Kang, M.R., Moon, E.Y., Jeon, Y.J.,
Gonzaga, E.R., 2009. Role of UV light in photoaging, skin aging, and skin cancer. Park, S.K., Kim, H.M., 2005. Estrogen receptor-independent inhibition of tumor
Am. J. Clin. Dermatol. 10 (Suppl. 1), 19–24. necrosis factor-alpha gene expression by phytoestrogen equol is mediated by
Gopaul, R., Knaggs, H., Lephart, E.D., 2012. Biochemical investigation and gene blocking nuclear factor-kappa B activation in mouse macrophages. Biochem.
analysis of equol: a plant and soy-derived isoflavonoid with anti-aging and Pharm. 71, 136–143, http://dx.doi.org/10.1016/j/bcp.2005.10.009.
anti-oxidant properties with potential human skin applications. Biofactors 38, Kang, N.J., Lee, K.W., Rogozin, E.A., Cho, Y.-Y., Heo, Y.-S., Bode, A.M., Lee, H.J., Dong,
44–52. Z., 2007a. Equol inhibits neoplastic cell transformation by targeting the
Gordon, J.R.S., Brieva, J.C., 2012. Unilateral dermatheliosis. N. Engl. J. Med. 366, e25, MEK/ERK/p90RSK/activator protein −1 pathway. J. Biol. Chem. 45,
http://dx.doi.org/10.1056/NEJMicm1104059. 32856–32866.
Gou, Z., Jiang, S., Zheng, C., Tian, Z., Lin, X., 2015. Equol inhibits LPS-induced Kang, J.S., Yoon, Y.D., Han, M.H., Han, S.D., Lee, K., Park, S.K., Kim, H.M., 2007b.
oxidative stress and enhances the immune response in chicken HD11 Equol inhibits nitric oxide production and inducible nitric oxide synthase gene
macrophages. Cell. Physiol. Biochem. 36, 611–621, http://dx.doi.org/10.1159/ expression through down-regulating the activation of Akt. Int.
000430124. Immunopharmacol. 7, 491–499, http://dx.doi.org/10.1016/j.intimp.2006.12.
Goukassian, D., Gad, F., Yaar, M., Eller, M.S., Nahal, U.S., Gilchrest, B.A., 2000. 004.
Mechanisms and implications of the age-associated decrease in DNA repair Khol, E., Steinbauer, J., Landthaler, M., Szeimies, R.M., 2011. Skin aging. J. Eur. Acad.
capacity. FASEB J. 14, 1325–1334. Dermatol. Venereol. 25, 873–884.
Greenwald, M.B.Y., Frusic-Zlotkin, M., Soroka, Y., Sasson, S.B., Bianco-Peled, H., King, R.A., Mano, M.M., Head, R.J., 1998. Assessment of isoflavonoid concentrations
Bitton, R., Kohen, R., 2015. Nitroxide delivery system for Nrf2 activation and in Australian bovine milk samples. J. Dairy Res. 65, 479–489, http://dx.doi.org/
skin protection. Eur. J. Pharm. Biopharm. 94, 123–134, http://dx.doi.org/10. 10.1017/S0022029998002891.
1016/j.ejpb.2015.05.008. Kligman, A.M., Koblenzer, C., 1997. Demographics and psychological implications
Gruber, F., Ornelas, C.M., Karner, S., Narzt, M.S., Nagelreiter, M.I., Gschwandtner, for the aging population. Dermatol. Clin. 15, 549–553.
M., Bochkov, V., Tschachler, E., 2015. Nrf2 deficiency causes lipid oxidation, Kohen, R., Nyska, A., 2002. Oxidation of biological systems: oxidative stress
inflammation, and matrix-protease expression in DHA-supplemented and phenomena, antioxidants, redox reactions, and methods for their
UVA-irradiated skin fibroblasts. Free Radic. Biol. Med. 88, 439–451, http://dx. quantification. Toxicol. Pathol. 30, 620–650, http://dx.doi.org/10.1080/
doi.org/10.1016/j/freeredbiomed.2015.05.006. 01926230290166724.
Gupta, S.C., Sundaram, C., Reuter, S., Aggarwal, B.B., 2010. Inhibiting NFkappaB Krahn-Bertil, E., Bolzinger, M.A., Andre, V., Orly, I., Kanitakis, J., Rousselle, P.,
activation by small molecules as a therapeutic strategy. Biochim. et Biophys. Damour, O., 2008. Expression of estrogen-related receptor gamma
Acta. 1799, 775–787, http://dx.doi.org/10.1016/k.bbagrm.2010.05.004. (ERRgamma) in human skin. Eur. J. Dermatol. 18, 427–432.
Halliwell, B., Gutteridge, J.M.C., 1999. Free Radicals in Biology and Medicine, 3rd Krishnamurthy, P., Wadhwani, A., 2012. Antioxidant Enzymes and Human Health,
edition. Oxford University Press, New York, pp. 617–783. chapter 1, In: El-Missiry, M.A. (Ed.), Antioxidant Enzyme- InTech Science,
Hamilton-Reeves, J.M., Vazque, G., Duval, S.J., Phipps, W.R., Kurzer, M.S., Messina, Technology & Medicine, Vienna, Austria, pp. 4–18. DOI: 10.5772/48109.
M.J., 2010. Clinical studies show no effects of soy protein or isoflavones on Kuwazuru, O., Saothong, J., Yoshikawa, Y., 2008. Mechanical approach to aging and
reproductive hormones in men: results of a meta-analysis. Fertil. Steril. 94, wrinkling of human facial skin based on the multistage buckling theory. Med.
997–1007, http://dx.doi.org/10.1016/j.fertnstert.2009.04.038. Eng. Phys. 30, 516–522.
Hayden, M.S., Ghosh, S., 2012. NF-kB, the first quarter-century: remarkable Lacher, S.E., Lee, J.S., Wang, X., Campbell, M.R., Bell, D.A., 2015. Beyond antioxidant
progress and outstanding questions. Genes Dev. 26, 203–234, http://dx.doi. genes in the ancient Nrf2 regulatory network. Free Radic. Biol. Med. 88,
org/10.1101/gad.183434.111. 452–465, 101016/j.freeradbiomed.2015.06.044.
Hellemans, L., Cortstjens, H., Neven, A., Declercq, L., Maes, D., 2003. Antioxidant Lee, M.J., Cha, J.H., Lim, K.M., Lee, O.-K., Bae, S., Kim, C.-H., Lee, K.-H., Lee, Y.N., Ahn,
enzyme activity in human stratum corneum shows seasonal variations with an K.J., An, S., 2015. Analysis of the microRNA expression profile of normal human
age-dependent recovery. J. Invest. Dermatol. 120, 434–439. dermal papilla cells treated with 5␣-dihydrotestosterone. Mol. Med. Rep. 12,
Hillebrand, G.G., 2010. Facial wrinkling: the marquee clinical sign of aging skin. In: 1205–1212, http://dx.doi.org/10.3892/mmr.2015.3478.
Farage, M.A., Miller, H.I., Maibach, H.I. (Eds.), Textbook of Aging Skin. Lephart, E.D., Summerfeldt, J.M., Andrus, M.B., 2014. Resveratrol: influences on
Springer-Verlag, Berlin, Heidelberg, pp. 911–918. gene expression in human skin. J. Funct. Foods 10, 377–384.
Hirvonen, J., Rajalin, A.M., Wohlfart, G., Adlercreutz, H., Wähälä, K., Aarnisalo, P., Lephart, E.D., 2009. Review Human Biology. Kendell/Hunt, Dubuque, Iowa, pp.
2011. Transcriptional activation of estrogen-related receptor gamma 68–94.
(ERRgamma) is stimulated by the phytoestrogen equol. J. Steroid Biochem. Lephart, E.D., 2013a. Protective effects of equol and their polyphenolic isomers
Mol. Biol. 123, 46–57, http://dx.doi.org/10.1016/j/jsbmb.2010.11.001. against dermal aging: microarray/protein evidence with clinical implications
Hoikkälä, A., Mustonen, A., Saastamoinen, S.I., Jokela, T., Taponen, J., Saloniemi, H., and unique delivery into human skin. Pharm. Biol. 51, 1391–1400.
Wähälä, K., 2007. High levels of equol in organic skimmed Finnish cow milk. Lephart, E.D., 2013b. Isoflavones and prenatal exposure to equol. In: Preedy, V.R.
Mol. Nutr. Food Res. 51, 782–786, http://dx.doi.org/10.1002/mnfr.200600222. (Ed.), Isoflavones: Chemistry, Analysis, Function. The Royal Society of
Hornebeck, W., 2003. Down-regulation of tissue inhibitor of matrix Chemistry, Thomas Graham House, Science Park, Cambridge, England, UK, pp.
metalloprotease-1 (TIMP-1) in aged human skin contributes to matrix 480–499.
degradation and impaired cell growth and survival. Pathol. Biol. 51, 569–573. Lephart, E.D., 2014a. Review: antioxidant and anti-aging properties of equol in
Hounsome, N., Hounsome, B., Deri Tomos, A., Edwards-Jones, G., 2009. Changes in prostate health (BPH). Open J. Endocr. Metab. Disord. 4, http://dx.doi.org/10.
anti-oxidant compounds in white cabbage during winter storage. Postharvest 4236/ojemd.2014.41001.
Biol. Technol. 52, 173–179. Lephart, E.D., 2014b. Safety assessment of R,S-equol as a dietary supplement for
Hounsome, H., Grail, B., Deri Tomos, A., Hounsome, B., Edwards-Jones, G., 2010. benign prostatic hyperplasia. Proc. Soc. Toxicol. pbn. 1585, p244.
High-throughput anti-oxidant profiling in vegetables by Fourier-transform ion Lephart, E.D., 2015. Modulation of aromatase by phytoestrogens. Enzyme Res.,
cyclotron resonance mass spectrometry. Funct. Plant Sci. Biotechnol. 4, 1–10. 2015, http://dx.doi.org/10.1155/2015/594656, article ID 594656.
E.D. Lephart / Ageing Research Reviews 31 (2016) 36–54 53

Li, H., Tsao, R., Deng, A., 2012. Factors affecting the antioxidant potential and health Pandel, R., Poljsak, B., Godic, A., Dahmane, R., 2013. Skin photoaging and the role of
benefits of plant foods. Can. J. Plant Sci. 92, 1101–1111, http://dx.doi.org/10. antioxidants in its prevention. ISRN Dermatol., http://dx.doi.org/10.1155/
4141/CJPS2011-239. 2013/930164, article ID 930164.
Lobo, V., Patil, A., Phatak, A., Chandra, N., 2010. Free radicals, antioxidants and Park, G., Oh, M.S., 2015. Acceleration of heat shock-induced collagen breakdown in
functional foods: impact on human health. Pharmacogn. Rev. 4, 118–126. human dermal fibroblasts with knockdown of NF-E2-related factor 2. BMB
Lund, T.D., Blake, C., Bu, L., Hamaker, A.N., Lephart, E.D., 2011. Equol an Rep. 48, 467–472, http://dx.doi.org/10.5483/BMBRep.2015.48.8.215.
isoflavonoid: potential for improved prostate health, in vitro and in vivo Park, E.-J., Pezzuto, J.M., 2015. The pharmacology of resveratrol in animals and
evidence. Reprod. Biol. Endocrinol. 9, 4, http://dx.doi.org/10.1186/1477-7827- humans. Biochim. Biophys. Acta 1852, 1071–1113.
9-4. Pelletier, G., Ren, L., 2004. Localization of sex steroid receptors in human skin.
Lundh, T.J.O., Petersson, H.I., Martinsson, K.A., 1990. Comparative levels of free and Histol. Histopathol. 19, 629–636.
conjugated plant estrogens in blood plasma of sheep and cattle fed oestrogenic Peng, Y., Xuan, M., Leung, V.Y.L., Cheng, B., 2015. Stem cells and aberrant signaling
silage. J. Agric. Food Chem. 38, 1530–1534, http://dx.doi.org/10.1021/ of molecular systems in skin aging. Ageing Res. Rev. 19, 8–21, http://dx.doi.
jf00097a022. org/10.1016/j.arr.2014.10.006.
Ma, Y., Sullivan, C., Schreihofer, D.A., 2010. Dietary genistein and equol (4 , 7 Phillips, T.J., Symons, J., Menon, S., 2008. HT Study Group. Does hormone therapy
isoflavandiol) reduce oxidative stress and protect rats against focal cerebral improve age-related skin changes in postmenopausal women? J. Am. Acad.
ischemia. Am. J. Physiol. Regul. Integr. Comp. Physiol. 299, R871–R877, http:// Dermatol. 59, 397–404.
dx.doi.org/10.1152/ajpregu.00031.2010. Pierard, G.E., Paquet, P., Xhauflaire-Uhoda, E., Quatresooz, P., 2010. Physiological
Madison, K.C., 2003. Barrier function of the skin: ‘la raison d’etre’ of the epidermis. variations during aging. In: Farage, M.A., Miller, K.W., Maibach, H.I. (Eds.),
J. Invest. Dermatol. 121, 231–241. Textbook of Aging Skin. Springer-Verlag, Berlin Heidelberg, pp. 45–54.
Maes, M., Galecki, P., Chang, Y.S., Berk, M., 2011. A review on the oxidative and Poljsak, B., Dehmane, R., 2012. Free radicals and extrinsic skin aging. Dermatol.
nitrosative (O&NS) pathways in major depression and their possible Res. Pract., http://dx.doi.org/10.1155/2012/135206, article ID: 135206.
contribution to the (neuro)degenerative process in that illness. Prog. Pomari, E., Valle, L.D., Pertile, P., Colombo, L., Thorton, M.J., 2015. Intracrine sex
Neuro-Psychpharm. Biol. Psychiatry 35, 676–692, http://dx.doi.org/10.1016/j. steroid synthesis and signaling in human epidermal keratinocytes and dermal
pnpbp.2010.05.004. fibroblasts. FASEB J. 29, 508–524, http://dx.doi.org/10.1096/fj.14-251363.
Magee, P.J., Raschke, M., Steiner, C., Duffin, J.G., Pool-Zobel, B.L., Jokela, T., Wähälä, Pusparini, Y., Hidayat, A., 2015. Effect of soy isoflavone supplementation on
K., Rowland, R.I., 2006. Equol: a comparison of the effects of the racemic endothelia dysfunction and oxidative stress in equol-producing
compound with that of the purified S-enantiomer on the growth, invasion, and postmenopausal women. Endocr. Metab. Immune Disord. Drug Targets 15,
DNA integrity of breast and prostate cells in vitro. Nutr. Cancer 54, 232–242. 71–79, http://dx.doi.org/10.2174/1871530314666141202123309.
Makrantonaki, E., Zouboulis, C.C., 2009. Androgens and ageing of the skin. Curr. Qin, Z., Voorhees, J.J., Fisher, G.J., Quan, T., 2014. Age-associated reduction of
Opin. Endocrinol. Diab. Obes. 16, 240–245, http://dx.doi.org/10.1097/MED. cellular spreading/mechanical force up-regulates matrix metalloproteinase-1
0b013e32832b71dc. expression and collagen fibril fragmentation via c-Jun/AP-1 in human dermal
Mann, G.E., Bonacasa, B., Ishii, T., Siow, R.C.M., 2009. Targeting the redox sensitive fibroblasts. Aging Cell 13, 1028–1037, 10.111.acel.12265.
Nrf2-Keep1 defense pathway in cardiovascular disease: protection afforded by Quan, C., Cho, M.K., Perry, D., Quan, T., 2015. Age-associated reduction of cell
dietary isoflavones. Curr. Opin. Pharmacol. 9, 139–145. spreading induces mitochondrial DNA common deletion by oxidative stress in
Marconi, A., Terracina, M., Fila, C., Franchi, J., Bonte, F., Romagnoli, G., Maurelli, R., human skin dermal fibroblasts: implication for human skin connective tissue
Failla, C.M., Dumas, M., Pincelli, C., 2003. Expression and function of aging. J. Biomed. Sci. 22, 62, http://dx.doi.org/10.1186/s12929-105-0167-6.
neurotrophins and their receptors in cultured human keratinocytes. J. Invest. Qunshan, J., Nash, J.F., 2010. Pathology of aging skin. In: Farage, M.A., Miller, K.W.,
Dermatol. 121, 1515–1521, http://dx.doi.org/10.1111/j.1523-1747.2003. Maibach, H.I. (Eds.), Textbook of Aging Skin. Springler-Verlag, Berlin
12624.x. Heidelberg, pp. 277–291.
Maritim, A.C., Sanders, R.A., Watkins III, J.B., 2003. Diabetes, oxidative stress, and Rabe, J.H., Mamelak, A.J., McElgunn, P.J., Morison, W.L., Sauder, D.N., 2006.
antioxidants: a review. J. Biochem. Mol. Toxicol. 17, 24–38, http://dx.doi.org/ Photoaging: mechanisms and repair. J. Am. Acad. Dermatol. 55, 1–19.
10.1002/jbt.10058. Ramara, A., Duckles, S.P., Krause, D.N., Proaccio, V., 2007. Estrogen suppresses brain
Markiewicz, M., Znoyko, S., Stawski, L., Ghatnekar, A., Gilkeson, G., Trojanowska, mitochondrial oxidative stress in female and male rats. Brain Res. 1176, 71–81.
M., 2013. A role for estrogen receptor-␣ and estrogen receptor-␤ in collagen Rhie, G., Shin, M.H., Seo, J.Y., Choi, W.W., Kim, K.H., Park, K.C., Eun, H.C., Chung, J.H.,
biosynthesis in mouse skin. J. Invest. Dermatol. 133, 120–127. 2001. Aging- and photoaging-dependent changes of enzymic and nonenzymic
Martini, F., 2004. Fundamentals of Anatomy and Physiology. Benjamin-Cummings, antioxidants in the epidermis and dermis of human skin in vitro. J. Invest.
San Francisco, CA. Dermatol. 117, 1212–1217.
Mazur, W., Adlercreutz, H., 2000. Overview of naturally occurring endocrine-active Rice-Evans, C., 2001. Flavonoid antioxidants. Curr. Med. Chem. 8, 797–807.
substances in the human diet in relation to human health. Nutrition 16, Rittie, L., Fisher, G.J., 2002. UV-light-induced signal cascades and skin aging. Ageing
654–658. Res. Rev. 1, 705–720.
Meadows, C., Moore, D.J., Moore, D.M., Draelos, Z.D., Kern, D., 2014. Age-related Robinson, J.K., Amonette, R., Wyatt, S.W., Bewerse, B.A., Bergfeld, W.F., Farris, P.K.,
NADH oxidase (arNOX)-catalyzed oxidative damage to skin protein. Arch. 1998. Executive summary of the national ‘Sun safety: protecting our future’
Dermatol. Res. 306, 645–652, http://dx.doi.org/10.1007/s00403-014-1472-8. conference. American Academy of Dermatology and Centers for Disease
Menon, G.K., 2002. New insights into skin structure: scratching the surface. Adv. Control and Prevention. J. Am. Acad. Dermatol. 38, 774–780.
Drug Deliv. Rev. 54, S3–S17. Rothwell, J.A., Day, A.J., Morgan, M.R., 2005. Experimental determination of
Mera, S.L., Lovell, C.R., Russell Jones, R., Davies, J.D., 1987. Elastic fibers in normal octanol-water partition coefficients of quercetin and related flavonoids. J.
and sun-damaged skin: an immunohistochemical study. Br. J. Dermatol. 117, Agric. Food Chem. 53, 4355–4360.
21–27. Rowlands, D.J., Chapple, S., Siow, R.C.M., Mann, G.E., 2011. Equol-stimulated
Mitchell, J.H., Gardner, P.T., McPhail, D.B., Morrice, P.C., Collins, A.R., Duthie, G.C., mitochrondrial reactive oxygen species activate endothelial nitric oxide
1998. Antioxidant efficacy of phytoestrogens in chemical and biological model synthase and redox signaling in endothelial cells, roles for F-actin and GPR30.
systems. Arch. Biochem. Biophys. 360, 142–148. Hypertension 57, 833–840.
Natarajan, V.T., Ganju, P., Ramkumar, A., Grover, R., Gokhale, R.S., 2014. Rufer, C.E., Kulling, S.E., 2006. Antioxidant activity of isoflavones and their major
Multifaceted pathways protect human skin from UV radiation. Nat. Chem. Biol. metabolites using different in vitro assays. J. Agric. Food Chem. 54, 2926–2931.
10, 542–551, http://dx.doi.org/10.1038/nchembio.1548. Schreck, R., Rieber, R., Baeuerle, P.A., 1991. Reactive oxygen intermediates as
National Institutes of Health, Office of Dietary Supplements, Vitamin D, 2016. apparently widely used messengers in the activation of the NF-kB
https/ods.od.nih.gov/factsheets/vitamin-healthprofessional/#h4. transcription factor and HIV-1. EMBO J. 10, 2247–2258.
Naylor, E.C., Watson, R.E.B., Sherratt, M., 2011. Molecular aspects of skin aging. Schwartz, D., Mayaux, M.J., 1982. Female fecundity as a function of age: results of
Maturitas 69, 249–256, http://dx.doi.org/10.1016/j.maturitas.2011.04.011. artificial insemination in 2193 nulliparous women with azoospermic
Nithy, M., Suguna, S., Rose, C., 2003. The effect of nerve growth factor on the early husbands. N. Engl. J. Med. 306, 404–406.
responses during the process of wound healing. Biochim. Biophys. Acta 1620, Seite, S., Zucchi, H., Sepiter, D., Igondjo-Tchen, S., Senni, K., Godeau, G., 2006.
25–31. Elastin changes during chronological and photo-ageing: the important role of
Nitsch, S.M., Wittmann, F., Angele, P., Wichmann, M.W., Hatz, R., lysozyme. J. Eur. Acad. Dermatol. Venereol. 20, 980–987, http://dx.doi.org/10.
Hernandez-Richter, T., Chaudry, I.H., Jauch, K.W., Angele, M.R., 2004. 1111/j.1468-3083.2006.01706.x.
Physiological levels of 5 alpha-dihydrotesterone depress wound immune Sekhar, K.R., Freeman, M.L., 2015. Nrf2 promotes survival following exposure to
function and impair wound healing following trauma-hemorrhage. Arch. Surg. ionizing radiation. Free Radic. Biol. Med. 88, 268–274, http://dx.doi.org/10.
139, 157–163. 1016/j/freeredbiomed.2015.04.035.
Nkengne, A., Bertin, C., 2012. Aging and facial changes-documenting clinical signs: Seo, J.Y., Chung, J.H., 2006. Thermal aging: a new concept of skin aging. J. Dermatol.
part 1: clinical changes of the aging face. Skinmed 10, 284–289. Sci. Suppl. 2, S13–S22.
Ortman, J.M., Velkoff, V.A., Hogan, H., 2014. An Aging Nation: The Older Population Setchell, K.D.R., Clerici, C., 2010. Equol: history, chemistry, and formation. J. Nutr.
in the United States. US Bureau of the Census, May. 140, 1355S–1362S.
Oyama, A., Ueno, T., Uchiyama, S., Aihara, T., Miyake, A., Kondo, S., Matsunage, K., Setchell, K.D.R., Clerici, C., Lephart, E.D., Cole, S.J., Heenan, C., Castellani, D., Wolfe,
2012. The effects of natural S-equol supplementation on skin aging in B.E., Nechemias-Zimmer, L., Brown, N.M., Lund, T.D., Handa, R.J., 2005. S-Equol,
postmenopausal women: a pilot randomized placebo-controlled trail. a potent ligand for estrogen receptor {beta}, is the exclusive enantiomeric
Menopause 19, 202–210, http://dx.doi.org/10.1097/gme.0b013e318227427b. form of the soy isoflavone metabolite produced by human intestinal bacterial
flora. Am. J. Clin. Nutr. 81, 1072–1079.
54 E.D. Lephart / Ageing Research Reviews 31 (2016) 36–54

Setchell, K.D.R., Zhao, X., Jah, P., Heubi, J.E., Brown, N.M., 2009. The Warren, R., Gartstein, V., Kligman, A.M., Montagna, W., Allendorf, R.A., Riddler,
pharmacokinetic behavior of the soy isoflavone metabolite S-(−) equol and its G.M., 1991. Age, sunlight, and facial skin: a histologic and quantitative study. J.
diastereoisomer R-(+)equol in healthy adults determined by using Am. Acad. Dermatol. 25, 751–760.Weber, C., Podda, M., Rallis, M., Thiele, J.J.,
stable-isotope-labeled tracers. Am. J. Clin. Nutr. 90, 1029–1037. Traber, M.G., Packer, L., 1997. Efficacy of topically applied tocophenols and
Shindo, Y., Witt, E., Han, D., Epstein, W., Packer, L., 1994. Enzymic and non-enzymic tocotrenols in protection of murine skin from oxidative damage induced by
antioxidants in epidermis and dermis in human skin. J. Invest. Dermatol. 102, UV-irradiation. Free Radic. Biol. Med. 22, 761–769.
122–124. Wend, K., Wend, P., Krum, S.A., 2012. Tissue-specific effects of loss of estrogen
Spiro, A., Buttriss, J.L., 2014. Vitamin D: an overview of vitamin D status and intake during menopause and aging. Front. Endocrinol. (Lausanne) 3, 1–19.
in Europe. Nutr. Bull. 39, 322–350. Widyarini, S., Domanski, D., Painter, N., Reeve, V.E., 2006a. Estrogen receptor
Strickland, I., Rhodes, L.E., Flanagan, B.F., Friedmann, P.S., 1997. TNF␣ and IL-8 are signaling protects against immune suppression by UV radiation exposure.
upregulated in the epidermis of normal human skin after UVB exposure: Proc. Natl. Acad. Sci. U. S. A. 103, 12837–12842, http://dx.doi.org/10.1073/pnas.
correlation with neurtrophil accumulation and E-selection expression. J. 0603642103.
Invest. Dermatol. 108, 763–768. Widyarini, S., Allanson, M., Gallagher, N.L., Pedley, J., Boyle, G.M., Parsons, P.G.,
Takahashi, Y., Moriwaki, S., Sugiyama, Y., Yamazaki, Y.E.-K., Mori, T., Takigawa, M., Whiteman, D.C., Walker, C., Reeve, V.E., 2006b. Isoflavonoid photoprotection in
2005. Decreased gene expression responsible for post-ultraviolet DNA repair mouse and human skin is dependent on metallothionein. J. Invest. Dermatol.
synthesis is aging: a possible mechanism of age-related reduction in DNA 126, 198–204, http://dx.doi.org/10.1038/sj.jid.5700013.
repair capacity. J. Invest. Dermatol. 124, 435–442. Widyarini, S., Domanski, D., Painter, N., Reeve, V.E., 2012. Photoimmune protective
Thiele, J.J., Traber, M.G., Packer, L., 1998. Depletion of human stratum corneum effect of the phytoestrogenic isoflavonoid equol is partially due to its
vitamin E: an early and sensitive in vivo marker of UV induced antioxidant activities. Photochem. Photobiol. Sci. 11, 1186–1192, http://dx.doi.
photo-oxidation. J. Invest. Dermatol. 110, 756–761. org/10.1039/c2pp25022e.
Thornton, M.J., Taylor, A.H., Mulligan, K., Al-Azzawi, F., Lyon, C.C., O’Driscoll, J., Xing, M.H., Hui, X., Zhong, W., 2009. In vitro human topical bioactive drug
Messenger, A.G., 2003. Oestrogen receptor beta is the predominant oestrogen transdermal absorption: estradiol. Cutan. Ocul. Toxicol. 28, 171–175.
receptor in human scalp skin. Exp. Dermatol. 12, 181–190. Yaar, M., Grossman, K., Eller, M., Gilchrest, B.A., 1991. Evidence for nerve growth
Tostes, R.C., Carneiro, F.S., Carvalho, M.H.C., Reckelhoff, J.F., 2016. Reactive oxygen factor-mediated paracrine effects in human epidermis. J. Cell Biol. 115,
species: players in the cardiovascular effects of testosterone. Am. J. Physiol. 821–828.
Regul. Integr. Comp. Physiol. 310, R1–R14. Yoon, G.-A., Park, S., 2014. Antioxidant action of soy isoflavones on oxidative stress
Tulah, A.S., Birch-Machin, M.A., 2013. Stressed out mitochondria: the role of and antioxidant enzyme activities in exercised rats. Nutr. Res. Pract. 8,
mitochondria in ageing and cancer focusing on strategies and opportunities in 618–624.
human skin. Mitochondrion 13, 444–453, http://dx.doi.org/10.1016/j.mito. Zhai, S., Yarr, M., Doyle, S.M., Gilchrest, B.A., 1996. Nerve growth factor rescues
2012.11.007. pigment cells from ultraviolet-induced apoptosis by upregulating BCL-2 levels.
Uitto, J., 2008. The role of elastin and collagen in cutaneous aging: intrinsic aging Exp. Cell Res. 224, 335–343.
versus photoexposure. J. Drugs Dermatol. 7, S12–S17. Zhang, T., Liang, X.Y., Shi, L.Y., Wang, L., Chen, J.L., Kang, C., Zhu, J.D., Mi, M.T., 2013.
Valko, M., Leibfritz, D., Moncol, J., Mazur, M.T., Telser, J., 2007. Free radicals and Estrogen receptor and PI3K/Akt signaling pathway involvement in
antioxidants in normal physiological functions and human disease. Int. J. S-equol-induced activation of Nrf2/ARE in endothelia cells. PLoS One 8 (2013),
Biochem. Cell Biol. 39, 44–84. http://dx.doi.org/10.1371/journal.pone.0079075.
Velarde, M.C., Flynn, J.M., Day, N.U., Melvo, S., Campisi, J., 2012. Mitochondrial Zhong, W., Xing, M.M.Q., Hui, X., Maibach, H.I., 2009. A stochastic model for
oxidative stress caused by SOD deficiency promotes cellular senescene and transepidermal drug delivery. Skin Res. Technol. 15, 407–411.
aging phenotypes in the skin. Aging 4, 3–12. Zouboulis, C.C., Makrantonaki, E., 2011. Clinical aspects and molecular diagnostics
Walker, S.L., Hawk, J.L.M., Young, A.R., 2003. Acute and chronic effects of ultraviolet of skin aging. Clin. Dermatol. 29, 3–14, http://dx.doi.org/10.1016/j.
radiation on the skin. In: Freedberg, I.M., Eisen, A.Z., Wolff, K.F., Austen, L.A., clindermatol.2010.07.001.
Goldsmith, L.A., Katz, S.I. (Eds.), Fitzpatrick’s Dermatology in General Medicine.
, 6th edition. McGraw-Hill, New York, pp. 1275–1282.
Wang, S., Moustaid-Moussa, N., Chen, L., Mo, H., Shastri, A., Su, R., Bapat, P., Kwun,
I., Shen, C.-L., 2014. Novel insights of dietary polyphenols and obesity. J. Nutr.
Biochem. 25, 1–18.

You might also like