10,357
Views
136
CrossRef citations to date
0
Altmetric
Review

Estrogens and aging skin

Pages 264-270 | Received 07 Aug 2012, Accepted 04 Feb 2013, Published online: 01 Apr 2013

Abstract

Estrogen deficiency following menopause results in atrophic skin changes and acceleration of skin aging. Estrogens significantly modulate skin physiology, targeting keratinocytes, fibroblasts, melanocytes, hair follicles and sebaceous glands, and improve angiogenesis, wound healing and immune responses. Estrogen insufficiency decreases defense against oxidative stress; skin becomes thinner with less collagen, decreased elasticity, increased wrinkling, increased dryness and reduced vascularity. Its protective function becomes compromised and aging is associated with impaired wound healing, hair loss, pigmentary changes and skin cancer.

 

Skin aging can be significantly delayed by the administration of estrogen. This paper reviews estrogen effects on human skin and the mechanisms by which estrogens can alleviate the changes due to aging. The relevance of estrogen replacement, selective estrogen receptor modulators (SERMs) and phytoestrogens as therapies for diminishing skin aging is highlighted. Understanding estrogen signaling in skin will provide a basis for interventions in aging pathologies.

Aging and Estrogen Deficiency

Multiple endocrine changes are associated with aging, but the best example of programmed aging in mammals is demonstrated by aging in the female reproductive system. The menopause is the result of a transition from full ovarian function to a complete lack of ovarian estrogen biosynthesis occurring in women around the age of 50 years old. Since Allen and Doisy first identified estrogen as a hormone in 1923,Citation1 its importance in female reproductive tissues has been well established. However, in recent years, this time-honored concept of estrogen as a female sex hormone has significantly altered; it is now apparent that estrogens have additional important and diverse functions in a plethora of tissues in both sexes, including the bone, brain, skeletal muscle, adipose tissue, colon, vascular system and skin; the loss of estrogen with aging has detrimental effects on these tissues, resulting in osteoporosis and an increased risk of cardiovascular disease.Citation2

Mechanism of Estrogen Action

The principle source of estrogen biosynthesis in females of reproductive age is the ovary. In men, estradiol can be produced in peripheral tissues by the actions of aromatase on androstenedione and testosterone.Citation3 Humans are unusual in that the adrenal cortex secretes large quantities of a precursor androgen, dehydroepiandrosterone (DHEA); following conversion into active steroids in peripheral tissues it provides the main source of active estrogens in post-menopausal women.Citation4 Even so, secretion of DHEA reduces with aging, with levels as low 10–20% of the peak concentrations in the elderly, so peripheral estrogen biosynthesis is also significantly reduced.Citation4 Estrogens can signal in a multifaceted manner involving diverse receptors that modulate genomic or non-genomic pathways, which in turn may have independent, synergistic, or opposing actions. In addition, cell specific co-factors are also required and ligands that display estrogenic activity in some cells, paradoxically exhibit estrogen antagonism in others.Citation5

Classical Mechanism of Action: Genomic Signaling

Two related, but distinct, tissue-dependent intracellular estrogen receptors (ERα and ERβ) have been identified as members of the superfamily family of nuclear hormone receptors.Citation5,Citation6 The ERα and ERβ proteins share approximately 97% homology in the DNA binding domain, with only a few amino acids differing in this region. However, in the ligand binding domain they only share 59% homology, while they share little homology in other domains.Citation7 With such a difference in the ligand binding domain, it could be anticipated that the receptors would bind estradiol with different affinities; this however is not the case, since 17β-estradiol has a similar affinity for both receptors.

ERα and ERβ are ligand activated nuclear transcription factors that enhance target-gene transcription upon binding to chromatin. Activation of the target gene by 17β-estradiol initiates an increase in the transcriptional activities via interaction with specific DNA palindrome estrogen response elements (ERE), found in the promoter region of estrogen-regulated target genes.Citation8 The recruitment of a large coactivator complex composed of p160 coactivators including GRIP1 and SRC-1 and the histone acetyltransferases p300/CREB-binding protein and pCAF are also required.Citation9 While identification of ERE is mediated by the DNA binding domain, the mediation of coactivator recruitment occurs via distinct activation functions (AF) located in the N-terminal domain (AF-1) and the ligand binding domain (AF-2). Coactivators are tissue-specific and ERα and ERβ can have diverse requirements for coactivators in a cell and tissue dependent manner.Citation10 In addition, estrogen receptors may also interact with other transcription factors bound to related DNA binding sites through protein-protein interactions. One such example is that either ERα or ERβ can augment the transcription of genes that contain AP-1 sites, the related binding site for the Jun/Fos complex.Citation11 When ERα and ERβ are co-localized within a cell, they can form both homodimers and heterodimers, although evidence suggests that ERα homodimers and ERα/ERβ heterodimers are formed in preference to ERβ homodimers, therefore the response of the cell to estrogen may be determined by the ratio of ERα:ERβ.Citation12

Estrogen Receptor: Non-Genomic Signaling

While the best described mechanism of estrogen signaling is mediated via the nuclear receptor proteins ERα and ERβ, it is now apparent that many cells also express membrane estrogen receptors coupled to cytosolic signal transduction proteins that direct signaling cascades via conventional second messengers including adenylate cyclase, cAMP, phospholipase C, protein kinase C, and the mitogen-activated protein kinase (MAPK) producing rapid responses to estrogen.Citation13 Although a number of cell-based studies have suggested G-protein-coupled receptors (GPCR), most notably GPR30 act as cell membrane estrogen receptors, there are few in vivo studies that can provide evidence to support this.Citation14

On the other hand, the presence of the classical ERα nuclear receptor has frequently been localized at the cell membrane. Membrane-bound classical ERs appear to work in partnership with GPCRs such as GPR30 located in the cell membrane to transmit rapid signals.Citation15 Verification that classical ERs are trafficked to the cell membrane following palmitoylation in the cytoplasm is now emerging.Citation16 Palmitoylation of monomeric ER encourages a physical interaction with the caveolin-1 protein required to help transport ER to the caveolae rafts in the cell membrane.Citation14 Although membrane-bound classical ERs lack the structural signaling domains of tyrosine kinase receptors, there is increasing evidence that they can also transactivate the EGF or IGF-1 receptors to stimulate kinase cascades.Citation17 For example, if the IGF-1 signaling pathway is blocked, uterine cells are unresponsive to estradiol while in ERα knockout cells, the effect of IGF-1 on proliferation and gene transactivation is lost.Citation18 Furthermore, the neuroprotective effects of estradiol are lost in the presence of a specific IGF- 1 receptor antagonist, while estrogen receptors are necessary for IGF-1 dependent neuronal differentiation and survival.Citation19 Complex interactions between estrogen and IGF-1 signaling pathways have also recently been reported in cultured human skin fibroblasts and the human sebocyte cell line.Citation20 Furthermore, in ovariectomized mice IGF-1 has been shown to improve wound healing, while it has no effect in ERα null mice.Citation21 The same study, using specific receptor antagonists, demonstrated that while the effects of IGF-1 on re-epithelialisation were mediated via the IGF-1 receptor, the anti inflammatory effects of IGF-1 were mediated via ERα, providing further support for the complexity of estrogen signaling in the skin.

The Menopause and Skin Aging

Following menopause many women detect a swift commencement of skin aging; skin becomes thinner with decreased collagen content, decreased elasticity, increased wrinkling and increased dryness. Many of these effects can be reversed by estrogen replacement which increases epidermal hydration, skin elasticity and skin thickness as well as reducing skin wrinkles and augmenting the content and quality of collagen and the level of vascularisation.Citation5,Citation6,Citation13 A recent immunohistochemical study has shown that while there is no difference in the expression of ERα and ERβ between male and female human skin, the expression of ERβ is significantly decreased in the epidermis of those above 70 years of age.Citation22

Many of the effects of estrogen on aging human skin have been derived by comparing post-menopausal women taking estrogen replacement therapy with women who have not. An increase in epidermal thickness in human female skin following six months of oral estrogen administration has been reportedCitation23 with an increase in keratinocyte volume and more defined rete ridges.Citation24 A study of elderly males and females has confirmed that administration of topical estrogen increases keratinocyte proliferation and epidermal thickness after only two weeks.Citation25 In estrogen deficient women skin thickness is reduced by 1.13% and collagen content by 2% per postmenopausal year.Citation26 Type I and III skin collagen is thought to decrease by as much as 30% in the first five years after menopause,Citation27,Citation28 which parallels the reduction in bone mass observed in post-menopausal women.Citation26 This decrease in skin thickness and collagen content in elderly females correlates with the period of estrogen deficiency rather than chronological age.Citation26-Citation28 A difference in collagen subtypes has also been recognized in post-menopausal women; compared with pre-menopausal women, post-menopausal women demonstrate a decrease in collagen types I and III and a reduction in the type III/type I ratio within the dermis, again correlating with the period of estrogen deficiency rather than chronological age.Citation28

A randomized, double-blind, placebo-controlled trial, has demonstrated that one year of oral estrogen therapy can increase dermal thickness by 30% in post-menopausal women,Citation24 while a similar trial established that six months of treatment with oral estrogen increases skin collagen by 6.49%.Citation29 Other studies have reported an increase specifically in collagen type III.Citation30,Citation31 The increase in response to estrogen therapy in skin collagen content appears to be related to the initial collagen content, since in women with low skin collagen content, initially estrogen is therapeutic, but later becomes purely prophylactic, while in women with only mild collagen loss in the early years following menopause, the effect of estrogen is essentially prophylactic.Citation26 The topical administration of estrogen can also increase skin collagen as measured by increased levels of type I and type III procollagen.Citation32 However, with topical administration, the effect of estrogen is confined to the area where it was directly applied.Citation31,Citation32 Topical estradiol applied to the buttock skin of elderly males and females significantly increased type I procollagen in both sexes, although the increase was significantly higher in women.Citation25 The same study also demonstrated that estradiol increased tropoelastin and fibrillin, which may be associated with an increase in elastic fibers. In addition, estrogen also increased TGF-β and TGF-β type II receptor expression, which may be related to dermal fibroblast proliferation and extracellular matrix (ECM) secretion, while it downregulated the expression of matrixmetalloprotease-I (MMP-1), which may explain the increased collagen content seen in estrogen treated skin.Citation25

Skin wrinkling is synonymous with aging, but may also be a result of environmental and hormonal factors. Wrinkling is caused by decreased skin elasticity due to elastic degeneration and loss of connective tissue.Citation33 In early postmenopausal women, skin elasticity can decrease by 1.5% per year, in contrast to women administered topical estrogen which thickens elastic fibers, increases the number of fibers and improves their orientation in the papillary dermis.Citation34 When women who were at least five years post-menopause and had taken oral estrogen continuously, were compared with women who had never had estrogen replacement, the average wrinkle score was significantly lower in the estrogen-treated group, providing further evidence of the long-term benefits of estrogen therapy on detrimental skin changes associated with aging.Citation35

Estrogens and Wound Healing

Aging is associated with impaired wound healing which leads to challenging non-healing chronic wounds. The recognition of the importance of estrogen in skin physiology would suggest it may also have an important role in wound healing. A number of studies have provided evidence that estrogens have a role in all phases of wound healing by modifying the inflammatory response, accelerating re-epithelialisation, stimulating granulation tissue formation and regulating proteolysis.Citation36 Case-cohort studies on venous ulceration and pressure ulcers in elderly women reported that women over the age of 65 years taking estrogen replacement were less likely to develop venous ulceration (age-adjusted relative risk 0.65) or a pressure ulcer (age-adjusted relative risk 0.68) than those who were not.Citation37

The inflammatory phase

Estrogen receptors have been identified in human leucocytes, monocytes, macrophages and megakaryocytes and there is evidence to suggest that estrogens can affect the function of inflammatory cells. In human subjects treated with topical estrogen a reduced number of neutrophils were seen at the wound site seven days post-wounding.Citation38 Furthermore, estrogen altered the expression of neutrophil adhesion molecules and downregulated the expression of L-selectin, thereby reducing the ability of neutrophils to localize to sites of inflammation.Citation38 Impaired wound healing associated with aging is coupled with excessive neutrophil recruitment and protease productionCitation39 and reduced fibronectin levels.Citation40 Since estrogen therapy reduces the number of wound neutrophils it follows that indirectly it should increase wound fibronectin levels. Ashcroft and colleaguesCitation38 demonstrated that estrogen therapy was associated with reduced elastase activity and less degradation of fibronectin in human wound tissue. Estrogen has also been shown to downregulate the expression of macrophage migration inhibition factor (MIF),Citation41 a pro-inflammatory cytokine released by monocytes, T lymphocytes, endothelial cells and keratinocytes.

The proliferative phase

The proliferative phase of wound healing involves re-epithelialisation, angiogenesis, formation of granulation tissue and wound contraction. A comparison of wound healing in pre- and post-menopausal women identified delayed re-epithelialisation in the post-menopausal group, which was reversed in women administered with replacement estrogen.Citation42 The application of topical estrogen patches immediately prior to, and following wounding, has also been shown to increase the rate of re-epithelialisation in both sexes, coupled with a reduction in wound size.Citation38

In the wounds of post-menopausal estrogen-deficient women, reduced collagen deposition was also demonstrated when compared with young women, while in post-menopausal women taking estrogen replacement, wound healing was comparable to that of the younger age group.Citation42 Furthermore, in elderly males and females treated with topical estrogen prior to wounding, increased collagen levels were demonstrated at day seven post-wounding.Citation38 Interestingly, females deposited more collagen than their male counterparts.

The dermal fibroblast is the key mesenchymal cell involved in wound healing, expressing both ERα and ERβ43. Estrogen stimulates the migration of cultured human dermal fibroblasts derived from scalp,Citation43 breast,Citation44 and abdominal skin.Citation45 Interestingly, increased migration occurred only in response to 17β-estradiol and an ERα agonist, while an ERβ agonist had no effect.Citation45 The importance of ERα was highlighted further by the observation that migration in the presence of the ERα agonist was higher than that seen with 17β-estradiol alone.Citation45 Since a significant acceleration of cell migration was seen as early as 4 h, this suggests a non-genomic signaling pathway, perhaps via a cell membrane ERα.

TGF-β1 plays a key role in wound healing and in vivo, TGF-β1 expression is decreased in wounds of elderly females compared with younger counterparts, a reduction reversed by estrogen replacement,Citation42 suggesting that estrogen may indirectly influence granulation tissue formation by altering cytokine profiles within healing wounds. In vitro studies of human dermal fibroblasts have demonstrated an increase in TGF-β1 secretion in response to estradiol.Citation44 When the fibroblast monolayers were mechanically wounded in culture, this also stimulated secretion of TGF-β1, but paradoxically, estradiol inhibited the secretion of TGF-β1 in the mechanically wounded dermal fibroblasts.Citation44

The remodelling phase

The remodelling phase of wound healing relies on a controlled balance between synthesis and degradation of the ECM, with estrogen thought to influence both. In humans, estrogen is also associated with an overall increase in collagen deposition during the remodelling phase.Citation38,Citation42 This suggests estrogen may affect the balance between collagen synthesis and degradation and studies have demonstrated that although aging is associated with an increase in MMP expression, specifically MMP-2 and MMP-9, staining for MMP-9 was most obvious in elderly females, suggesting that the reduction in estrogen that occurs postmenopausally may affect proteinase production.Citation40

Estrogen and the Hair Cycle

Estrogens significantly inhibit hair growth in a number of mammalian species, but regulation of the hair cycle in humans by estrogens appears more complex.Citation46 In vivo, estradiol prolongs the anagen phase of the hair follicle, which is also evident during pregnancy when an increase in the number of hairs in anagen is seen.Citation47 Postpartum, these additional anagen follicles enter telogen, which causes increased hair loss and a temporary thinning of the hair. Limited trichogram evidence suggests that estrogens decrease the telogen rate and prolong the anagen phase when used to treat female pattern hair loss.Citation6 More recently it has been demonstrated that female pattern hair loss is linked to polymorphisms of the ERβ gene.Citation48 Further evidence comes from the use of aromatase inhibitors, which prevent the synthesis of estrogens; in these women a common treatment-related side-effect is scalp hair thinningCitation6 and a recent study has described a link between the risk of female pattern hair loss and a polymorphism of the gene encoding aromatase.Citation49 However, the response to estrogens by human hair follicles in vitro appears to show differences in terms of gender and site.Citation46

In situ, immunohistochemical studies have shown that in contrast to ERα, ERβ is strongly expressed in human non-balding scalp anagen hair follicles derived from both men and women.Citation50,Citation51 A more recent study using quantitative real-time RT-PCR has demonstrated that the expression of ERβ transcripts is significantly higher than the expression of ERα transcripts in cultured human dermal papilla, dermal sheath and dermal fibroblasts derived from female scalp.Citation43

Estrogens and Skin Pigmentation

In humans, hyperpigmentation has been documented during pregnancy (melasma), in women ingesting oral contraceptives containing estrogens and in female and male infants treated with ointments containing estrogen.Citation52 Particular regions of the body seem to be affected such as the genitals, abdomen, linea alba, face and mammary areola.Citation5 These clinical observations suggest that melanocytes can respond to estrogens by increasing their levels of pigmentation. However, the precise effect of estrogens on human melanocyte and melanoma biology remains controversial, and is exacerbated by a significant lack of information on the relative expression of estrogen receptors in both human melanocytes and melanomas.Citation5

Tyrosinase is the rate limiting enzyme in melanogenesis, catalyzing the conversion of l-tyrosine to 3,4-dihydroxyindole (DOPA), DOPA to DOPAquinone and subsequently DOPAquinone to 5,6- dihydroxyindole to indole-5, 6-quinone, which polymerises to produce melanin.Citation53 Therefore, tyrosinase activity can be determined by DOPA oxidase activity. Studies using proliferating melanocyte cultures showed that the M-box of Dopachrome tautomerase (DCT), a member of the tyrosinase gene family that includes the MITF CATGTG binding motif sequence, overlaps with the ERα binding element.Citation54 Proliferating melanocytes contained these MITF and ERα complexes, while in senescent cells only ERα complexes were found. These researchers also reported that MITF, together with ERα and the histone acetyltransferase p300, can synergistically induce high levels of DCT gene transcription in normal proliferating melanocytes. Collectively, these results suggest a mechanism for estrogens to directly regulate the DCT gene, leading to hyperpigmentation as seen in some pigmentation disorders associated with elevated levels of estrogens.

Non-Melanoma Skin Cancer

Interestingly, men exceed women in terms of incidence and mortality for basal cell carcinomas (BCC) and squamous cell carcinomas (SCC), with the incidence of non-melanoma skin cancer 2-fold higher in men compared with womenCitation55 and mortality rates from SCC higher in men than women.Citation56 Furthermore, of patients with a prior skin cancer, men have a 50% greater risk of developing a new BCC and a 3-fold higher risk of developing a new SCCCitation57 suggesting that women have some protection.

Aging Skin and Oxidative Stress

One of the hallmarks associated with chronological skin aging is an increase in inflammation. Premature skin aging or photoaging due to UV exposure induces chronic low grade inflammation which damages the skin by increasing the expression of proinflammatory cytokines and MMPs leading to detrimental changes.Citation58 In normal skin, cellular mitochondrial metabolism produces reactive oxygen species (ROS). The presence of antioxidant enzymes such as superoxide dismutase (SOD) maintains normal levels of ROS homeostasis and minimizes the level of cellular stress. Both UV exposure and inflammation result in elevated ROS and oxidative stress, increasing damage to DNA, proteins and lipids and lead to premature aging.Citation59

Estrogens have been demonstrated to have cytoprotective effects in a number of cells and tissues, although their precise mechanism of action is unclear. Friedreich’s ataxia is an inherited autosomal recessive condition that results in the functional absence of the protein Frataxin.Citation60 Since Frataxin is responsible for preventing the formation of ROS, its absence contributes to the development of a wide range of neurological disorders such as Alzheimer, Parkinson and Huntington disease.Citation61 Dermal fibroblasts derived from the skin of these individuals are extremely sensitive to free radical damage and oxidative stress. A recent study has demonstrated that estrogens can protect against oxidative stress induced in these fibroblasts, but the mechanism appears to be independent of the intracellular receptors ERα, ERβ or GPR30.Citation60 There is increasing evidence to suggest that the antioxidant property of estrogen is due to the presence of the A-ring phenol which can attenuate ROS created by the Fenton reaction by a cyclic phenol-quinol mechanism.Citation62 The same group went on to demonstrate that phenolic estrogenic compounds independent of any known ER can prevent oxidative damage to mitochondria in Friedreich’s ataxia cultured skin fibroblasts.Citation63 However, western blot analysis of these fibroblasts only demonstrated small amounts of ERβ, while the presence of ERα was not detected, and there was no comparison to ER levels in normal skin fibroblasts.Citation63 Normal human skin fibroblasts express mRNA transcripts and protein for both ERα and ERβ in culture, although quantification using real-time RT-PCR demonstrated that the expression of ERβ transcripts was approximately 20-fold higher than expression of ERα transcripts.Citation43 Exposure of cultured human primary keratinocytes and dermal fibroblasts to UV has shown that selective ERβ agonists significantly decreased inflammatory markers and MMPs, while an ERα selective agonist had no effect.Citation64

Selective Estrogen Receptor Modulators (SERMs)

Although estrogen replacement is effective at managing menopausal symptoms and preventing osteoporosis, the use of estrogens has also been implicated as a risk factor in breast and uterine cancer.Citation65 The recognition of the unusual properties of the non-steroidal triphenylethylene tamoxifen acing as an estrogen antagonist in some tissues e.g., mammary gland, or an estrogen agonist in others, led to the embodiment of the concept of selective estrogen receptor modulators or SERMs.Citation66

The drive to develop a SERM with an ideal agonist/antagonist profile has increased significantly over recent years. A SERM can bind to either ERα or ERβ, causing a conformational change in the receptor which will allow it to recruit essential co-factors, which may be co-activators or co-repressors, depending on either the tissue or the SERM. The complexes may modulate genes by either the classical ERE pathway or via protein-protein interactions.Citation66 This gives numerous combinations by which SERMS can modulate estrogen receptors in a tissue-specific manner.

However, despite the well documented effects of estrogen on skin physiology and aging, there is still very limited data on the effect of SERMs on the skin.Citation13 Raloxifene a SERM which is used successfully to prevent and treat postmenopausal osteoporosis, increases collagen biosynthesis in human skin fibroblastsCitation67 and a recent study has demonstrated that raloxifene has a similar effect to estrogen by increasing skin elasticity in postmenopausal women.Citation68

One of the fundamental aspects of cutaneous wound healing is an increase in dermal fibroblast proliferation, and recent studies in our laboratory have shown that monolayers of human dermal fibroblasts derived from peri-menopausal women demonstrate a significantly increased rate of proliferation in response to tamoxifen and raloxifene following mechanical wounding in vitro,Citation45 supporting the application of SERMs as potential therapeutic agents to improve wound healing in postmenopausal women.

Phytoestrogens

In the early 1920s, Bernard Zondek demonstrated that willow tree flowers mimicked estrogen, confirming the existence of plant estrogens or phytoestrogens. The phytoestrogens daidzein and genistein are naturally occurring isoflavones that are found in numerous edible plants, especially soybeans. The traditional Asian diet is renowned for being rich in soy containing phytoestrogens and epidemiological evidence first suggested the beneficial effects of a diet rich in estrogen-like compounds by comparing the health associated benefits of an Asian diet compared with a Western diet.Citation69 For example the rate of hip fracture in Asian populations is considerably less than those of whites residing in the United States.Citation70 Since these polyphenolic compounds can bind both ERα and ERβ and act as both estrogen agonists and antagonistsCitation71 they are considered to be naturally occurring SERMs and are potential contenders to provide a natural form of estrogen replacement in postmenopausal women.

Although in general phytoestrogens bind both ERα and ERβ, the isoflavones genistein and S-equol have a significantly greater affinity for ERβ and therefore can be categorised as ERβ selective agonists.Citation72 Both have positive effects on human skin; they can reduce UV-induced cell death in cultured keratinocytes, improve skin elasticity, reduce wrinkle depth and increase the production of type 1 procollagen.Citation73 More recently it has been reported that genistein offers protection against UV induced senescence in cultured human dermal fibroblasts by significantly upregulating intracellular SOD activity in a dose-dependent manner.Citation74

Another phytoestrogen, resveratrol found in grapes and red wine, can also activate ERα and ERβ and is a potent antioxidant with strong anti-inflammatory properties.Citation73 A recent study has demonstrated that resveratrol upregulates mitochondrial SOD in cultured human lung fibroblasts and human neuroblastoma cells.Citation75 Furthermore, the effect of resveratrol could be abolished by the ER antagonist ICI 182780. The effect of resveratrol on mitochondrial SOD could be replicated with either estradiol or an ERβ agonist, but not an ERα agonist, suggesting that resveratrol upregulates mitochondrial SOD via ERβ75.

Conclusions

The menopause causes hypoestrogenism, accelerating age-related deterioration of the skin. Estrogen administration has positive effects on human skin by delaying or preventing skin aging manifestations, but the use of estrogen replacement is a risk factor in breast and uterine cancer. Estrogens have important antioxidant properties, but estrogen signaling is complex and intricate, and estrogens can modulate a plethora of signaling pathways that are often cell or tissue-specific. In addition, there are an increasing number of synthetic compounds (SERMs) or naturally occurring compounds (phytoestrogens) that exhibit agonist or antagonist estrogenic properties, depending on the tissue. These could be ideal candidates to combat skin aging and other detrimental effects of hypoestrogenism, such as osteoporosis if we could exploit the positive effects of estrogen on human health, and avoid the negative aspects of estrogen signaling in tissues such as breast and uterus. Only a better understanding of the mechanisms of estrogen action and the structurally related phytoestrogens will help to identify possible candidates to delay the aging process.

References

  • Allen E, Doisy EA. An ovarian hormone preliminary report on its localisation, extraction and partial purification, and action in test animals. JAMA 1923; 81:819 - 21; http://dx.doi.org/10.1001/jama.1923.02650100027012
  • Wend K, Wend P, Krum SA. Tissue-specifc effects of loss of estrogen during menopause and aging. Front Endocrinol (Lausanne) 2012; 3:1 - 14; PMID: 22649402
  • Simpson ER. Genetic mutations resulting in estrogen insufficiency in the male. Mol Cell Endocrinol 1998; 145:55 - 9; http://dx.doi.org/10.1016/S0303-7207(98)00169-5; PMID: 9922099
  • Labrie F. Adrenal androgens and intracrinology. Semin Reprod Med 2004; 22:299 - 309; http://dx.doi.org/10.1055/s-2004-861547; PMID: 15635498
  • Thornton MJ. The biological actions of estrogens on skin. Exp Dermatol 2002; 11:487 - 502; http://dx.doi.org/10.1034/j.1600-0625.2002.110601.x; PMID: 12473056
  • Thornton MJ. Oestrogen functions in skin and skin appendages. Expert Opin Ther Targets 2005; 9:617 - 29; http://dx.doi.org/10.1517/14728222.9.3.617; PMID: 15948678
  • Gustafsson JA. Estrogen receptor beta--a new dimension in estrogen mechanism of action. J Endocrinol 1999; 163:379 - 83; http://dx.doi.org/10.1677/joe.0.1630379; PMID: 10588810
  • Klinge CM. Estrogen receptor interaction with estrogen response elements. Nucleic Acids Res 2001; 29:2905 - 19; http://dx.doi.org/10.1093/nar/29.14.2905; PMID: 11452016
  • Webb P, Nguyen P, Kushner PJ. Differential SERM effects on corepressor binding dictate ERalpha activity in vivo. J Biol Chem 2003; 278:6912 - 20; http://dx.doi.org/10.1074/jbc.M208501200; PMID: 12482846
  • Paech K, Webb P, Kuiper GG, Nilsson S, Gustafsson J, Kushner PJ, et al. Differential ligand activation of estrogen receptors ERalpha and ERbeta at AP1 sites. Science 1997; 277:1508 - 10; http://dx.doi.org/10.1126/science.277.5331.1508; PMID: 9278514
  • Smith CL, O’Malley BW. Coregulator function: a key to understanding tissue specificity of selective receptor modulators. Endocr Rev 2004; 25:45 - 71; http://dx.doi.org/10.1210/er.2003-0023; PMID: 14769827
  • Thornton MJ, Nelson LD, Taylor AH, Birch MP, Laing I, Messenger AG. The modulation of aromatase and estrogen receptor alpha in cultured human dermal papilla cells by dexamethasone: a novel mechanism for selective action of estrogen via estrogen receptor beta?. J Invest Dermatol 2006; 126:2010 - 8; http://dx.doi.org/10.1038/sj.jid.5700344; PMID: 16691199
  • Stevenson S, Thornton J. Effect of estrogens on skin aging and the potential role of SERMs. Clin Interv Aging 2007; 2:283 - 97; PMID: 18044179
  • Levin ER. Minireview: Extranuclear steroid receptors: roles in modulation of cell functions. Mol Endocrinol 2011; 25:377 - 84; http://dx.doi.org/10.1210/me.2010-0284; PMID: 20861220
  • Levin ER. G protein-coupled receptor 30: estrogen receptor or collaborator?. Endocrinology 2009; 150:1563 - 5; http://dx.doi.org/10.1210/en.2008-1759; PMID: 19307418
  • Pedram A, Razandi M, Sainson RC, Kim JK, Hughes CC, Levin ER. A conserved mechanism for steroid receptor translocation to the plasma membrane. J Biol Chem 2007; 282:22278 - 88; http://dx.doi.org/10.1074/jbc.M611877200; PMID: 17535799
  • Santen RJ, Fan P, Zhang Z, Bao Y, Song RX, Yue W. Estrogen signals via an extra-nuclear pathway involving IGF-1R and EGFR in tamoxifen-sensitive and -resistant breast cancer cells. Steroids 2009; 74:586 - 94; http://dx.doi.org/10.1016/j.steroids.2008.11.020; PMID: 19138696
  • Song RX, Fan P, Yue W, Chen Y, Santen RJ. Role of receptor complexes in the extranuclear actions of estrogen receptor alpha in breast cancer. Endocr Relat Cancer 2006; 13:Suppl 1 S3 - 13; http://dx.doi.org/10.1677/erc.1.01322; PMID: 17259556
  • Lebesgue D, Chevaleyre V, Zukin RS, Etgen AM. Estradiol rescues neurons from global ischemia-induced cell death: multiple cellular pathways of neuroprotection. Steroids 2009; 74:555 - 61; http://dx.doi.org/10.1016/j.steroids.2009.01.003; PMID: 19428444
  • Makrantonaki E, Vogel K, Fimmel S, Oeff M, Seltmann H, Zouboulis CC. Interplay of IGF-I and 17beta-estradiol at age-specific levels in human sebocytes and fibroblasts in vitro. Exp Gerontol 2008; 43:939 - 46; http://dx.doi.org/10.1016/j.exger.2008.07.005; PMID: 18755261
  • Emmerson E, Campbell L, Davies FC, Ross NL, Ashcroft GS, Krust A, et al. Insulin-like growth factor-1 promotes wound healing in estrogen-deprived mice: new insights into cutaneous IGF-1R/ERα cross talk. J Invest Dermatol 2012; 132:2838 - 48; http://dx.doi.org/10.1038/jid.2012.228; PMID: 22810305
  • Inoue T, Miki Y, Abe K, Hatori M, Hosaka M, Kariya Y, et al. The role of estrogen-metabolizing enzymes and estrogen receptors in human epidermis. Mol Cell Endocrinol 2011; 344:35 - 40; http://dx.doi.org/10.1016/j.mce.2011.06.015; PMID: 21740958
  • Punnonen R. Effect of castration and peroral estrogen therapy on the skin. Acta Obstet Gynecol Scand Suppl 1972; 21:3 - 44; PMID: 4511789
  • Maheux R, Naud F, Rioux M, Grenier R, Lemay A, Guy J, et al. A randomized, double-blind, placebo-controlled study on the effect of conjugated estrogens on skin thickness. Am J Obstet Gynecol 1994; 170:642 - 9; PMID: 8116726
  • Son ED, Lee JY, Lee S, Kim MS, Lee BG, Chang IS, et al. Topical application of 17beta-estradiol increases extracellular matrix protein synthesis by stimulating tgf-Beta signaling in aged human skin in vivo. J Invest Dermatol 2005; 124:1149 - 61; http://dx.doi.org/10.1111/j.0022-202X.2005.23736.x; PMID: 15955089
  • Brincat M, Versi E, Moniz CF, Magos A, de Trafford J, Studd JW. Skin collagen changes in postmenopausal women receiving different regimens of estrogen therapy. Obstet Gynecol 1987; 70:123 - 7; PMID: 3601260
  • Brincat M, Moniz CJ, Studd JW, Darby A, Magos A, Emburey G, et al. Long-term effects of the menopause and sex hormones on skin thickness. Br J Obstet Gynaecol 1985; 92:256 - 9; http://dx.doi.org/10.1111/j.1471-0528.1985.tb01091.x; PMID: 3978054
  • Affinito P, Palomba S, Sorrentino C, Di Carlo C, Bifulco G, Arienzo MP, et al. Effects of postmenopausal hypoestrogenism on skin collagen. Maturitas 1999; 33:239 - 47; http://dx.doi.org/10.1016/S0378-5122(99)00077-8; PMID: 10656502
  • Sauerbronn AV, Fonseca AM, Bagnoli VR, Saldiva PH, Pinotti JA. The effects of systemic hormonal replacement therapy on the skin of postmenopausal women. Int J Gynaecol Obstet 2000; 68:35 - 41; http://dx.doi.org/10.1016/S0020-7292(99)00166-6; PMID: 10687834
  • Savvas M, Bishop J, Laurent G, Watson N, Studd J. Type III collagen content in the skin of postmenopausal women receiving oestradiol and testosterone implants. Br J Obstet Gynaecol 1993; 100:154 - 6; http://dx.doi.org/10.1111/j.1471-0528.1993.tb15212.x; PMID: 8476808
  • Schmidt JB, Binder M, Demschik G, Bieglmayer C, Reiner A. Treatment of skin aging with topical estrogens. Int J Dermatol 1996; 35:669 - 74; http://dx.doi.org/10.1111/j.1365-4362.1996.tb03701.x; PMID: 8876303
  • Varila E, Rantala I, Oikarinen A, Risteli J, Reunala T, Oksanen H, et al. The effect of topical oestradiol on skin collagen of postmenopausal women. Br J Obstet Gynaecol 1995; 102:985 - 9; http://dx.doi.org/10.1111/j.1471-0528.1995.tb10906.x; PMID: 8652490
  • Shah MG, Maibach HI. Estrogen and skin. An overview. Am J Clin Dermatol 2001; 2:143 - 50; http://dx.doi.org/10.2165/00128071-200102030-00003; PMID: 11705091
  • Henry F, Piérard-Franchimont C, Cauwenbergh G, Piérard GE. Age-related changes in facial skin contours and rheology. J Am Geriatr Soc 1997; 45:220 - 2; PMID: 9033524
  • Wolff EF, Narayan D, Taylor HS. Long-term effects of hormone therapy on skin rigidity and wrinkles. Fertil Steril 2005; 84:285 - 8; http://dx.doi.org/10.1016/j.fertnstert.2004.12.062; PMID: 16084862
  • Emmerson E, Hardman MJ. The role of estrogen deficiency in skin ageing and wound healing. Biogerontology 2012; 13:3 - 20; http://dx.doi.org/10.1007/s10522-011-9322-y; PMID: 21369728
  • Margolis DJ, Knauss J, Bilker W. Hormone replacement therapy and prevention of pressure ulcers and venous leg ulcers. Lancet 2002; 359:675 - 7; http://dx.doi.org/10.1016/S0140-6736(02)07806-6; PMID: 11879867
  • Ashcroft GS, Greenwell-Wild T, Horan MA, Wahl SM, Ferguson MWJ. Topical estrogen accelerates cutaneous wound healing in aged humans associated with an altered inflammatory response. Am J Pathol 1999; 155:1137 - 46; http://dx.doi.org/10.1016/S0002-9440(10)65217-0; PMID: 10514397
  • Ashcroft GS, Lei KJ, Jin WW, Longenecker G, Kulkarni AB, Greenwell-Wild T, et al. Secretory leukocyte protease inhibitor mediates non-redundant functions necessary for normal wound healing. Nat Med 2000; 6:1147 - 53; http://dx.doi.org/10.1038/80489; PMID: 11017147
  • Ashcroft GS, Horan MA, Herrick SE, Tarnuzzer RW, Schultz GS, Ferguson MWJ. Age-related differences in the temporal and spatial regulation of matrix metalloproteinases (MMPs) in normal skin and acute cutaneous wounds of healthy humans. Cell Tissue Res 1997; 290:581 - 91; http://dx.doi.org/10.1007/s004410050963; PMID: 9369533
  • Ashcroft GS, Mills SJ, Lei KJ, Gibbons L, Jeong MJ, Taniguchi M, et al. Estrogen modulates cutaneous wound healing by downregulating macrophage migration inhibitory factor. J Clin Invest 2003; 111:1309 - 18; PMID: 12727922
  • Ashcroft GS, Dodsworth J, van Boxtel E, Tarnuzzer RW, Horan MA, Schultz GS, et al. Estrogen accelerates cutaneous wound healing associated with an increase in TGF-beta1 levels. Nat Med 1997; 3:1209 - 15; http://dx.doi.org/10.1038/nm1197-1209; PMID: 9359694
  • Stevenson S, Taylor AH, Meskiri A, Sharpe DT, Thornton MJ. Differing responses of human follicular and nonfollicular scalp cells in an in vitro wound healing assay: effects of estrogen on vascular endothelial growth factor secretion. Wound Repair Regen 2008; 16:243 - 53; http://dx.doi.org/10.1111/j.1524-475X.2008.00365.x; PMID: 18318810
  • Stevenson S, Nelson LD, Sharpe DT, Thornton MJ. 17beta-estradiol regulates the secretion of TGF-β by cultured human dermal fibroblasts. J Biomater Sci Polym Ed 2008; 19:1097 - 109; http://dx.doi.org/10.1163/156856208784909354; PMID: 18644234
  • Stevenson S, Sharpe DT, Thornton MJ. Effects of oestrogen agonists on human dermal fibroblasts in an in vitro wounding assay. Exp Dermatol 2009; 18:988 - 90; http://dx.doi.org/10.1111/j.1600-0625.2009.00864.x; PMID: 19469894
  • Ohnemus U, Uenalan M, Inzunza J, Gustafsson JA, Paus R. The hair follicle as an estrogen target and source. Endocr Rev 2006; 27:677 - 706; http://dx.doi.org/10.1210/er.2006-0020; PMID: 16877675
  • Lynfield YL. Effect of pregnancy on the human hair cycle. J Invest Dermatol 1960; 35:323 - 7; PMID: 13764567
  • Yip L, Zaloumis S, Irwin D, Severi G, Hopper J, Giles G, et al. Association analysis of oestrogen receptor beta gene (ESR2) polymorphisms with female pattern hair loss. Br J Dermatol 2012; 166:1131 - 4; http://dx.doi.org/10.1111/j.1365-2133.2011.10702.x; PMID: 22014031
  • Yip L, Zaloumis S, Irwin D, Severi G, Hopper J, Giles G, et al. Gene-wide association study between the aromatase gene (CYP19A1) and female pattern hair loss. Br J Dermatol 2012; 161:289 - 94; http://dx.doi.org/10.1111/j.1365-2133.2009.09186.x; PMID: 22014031
  • Thornton MJ, Taylor AH, Mulligan K, Al-Azzawi F, Lyon CC, O’Driscoll J, et al. Estrogen receptor beta (ERβ) is the predominant estrogen receptor in human scalp. Exp Dermatol 2003; 12:181 - 90; http://dx.doi.org/10.1034/j.1600-0625.2003.120209.x; PMID: 12702147
  • Thornton MJ, Taylor AH, Mulligan K, Al-Azzawi F, Lyon CC, O’Driscoll J, et al. The distribution of estrogen receptor beta is distinct to that of estrogen receptor alpha and the androgen receptor in human skin and the pilosebaceous unit. J Investig Dermatol Symp Proc 2003; 8:100 - 3; http://dx.doi.org/10.1046/j.1523-1747.2003.12181.x; PMID: 12895004
  • Beas F, Vargas L, Spada RP, Merchak N. Pseudoprecocious puberty in infants caused by a dermal ointment containing estrogens. J Pediatr 1969; 75:127 - 30; http://dx.doi.org/10.1016/S0022-3476(69)80113-7; PMID: 5790395
  • Schallreuter KU, Kothari S, Chavan B, Spencer JD. Regulation of melanogenesis--controversies and new concepts. Exp Dermatol 2008; 17:395 - 404; http://dx.doi.org/10.1111/j.1600-0625.2007.00675.x; PMID: 18177348
  • Schwahn DJ, Timchenko NA, Shibahara S, Medrano EE. Dynamic regulation of the human dopachrome tautomerase promoter by MITF, ER-alpha and chromatin remodelers during proliferation and senescence of human melanocytes. Pigment Cell Res 2005; 18:203 - 13; http://dx.doi.org/10.1111/j.1600-0749.2005.00229.x; PMID: 15892717
  • Preston DS, Stern RS. Nonmelanoma cancers of the skin. N Engl J Med 1992; 327:1649 - 62; http://dx.doi.org/10.1056/NEJM199212033272307; PMID: 1435901
  • Karagas MR, The Skin Cancer Prevention Study Group. Occurrence of cutaneous basal cell and squamous cell malignancies among those with a prior history of skin cancer. J Invest Dermatol 1994; 102:10S - 3S; http://dx.doi.org/10.1111/1523-1747.ep12386072; PMID: 8006426
  • Weinstock MA. Epidemiologic investigation of nonmelanoma skin cancer mortality: the Rhode Island Follow-Back Study. J Invest Dermatol 1994; 102:6S - 9S; http://dx.doi.org/10.1111/1523-1747.ep12385735; PMID: 8006441
  • Iddamalgoda A, Le QT, Ito K, Tanaka K, Kojima H, Kido H. Mast cell tryptase and photoaging: possible involvement in the degradation of extra cellular matrix and basement membrane proteins. Arch Dermatol Res 2008; 300:Suppl 1 S69 - 76; http://dx.doi.org/10.1007/s00403-007-0806-1; PMID: 17968569
  • Masaki H. Role of antioxidants in the skin: anti-aging effects. J Dermatol Sci 2010; 58:85 - 90; http://dx.doi.org/10.1016/j.jdermsci.2010.03.003; PMID: 20399614
  • Richardson TE, Yang SH, Wen Y, Simpkins JW. Estrogen protection in Friedreich’s ataxia skin fibroblasts. Endocrinology 2011; 152:2742 - 9; http://dx.doi.org/10.1210/en.2011-0184; PMID: 21540287
  • Simpkins JW, Dykens JA. Mitochondrial mechanisms of estrogen neuroprotection. Brain Res Rev 2008; 57:421 - 30; http://dx.doi.org/10.1016/j.brainresrev.2007.04.007; PMID: 17512984
  • Prokai L, Prokai-Tatrai K, Perjesi P, Zharikova AD, Perez EJ, Liu R, et al. Quinol-based cyclic antioxidant mechanism in estrogen neuroprotection. Proc Natl Acad Sci U S A 2003; 100:11741 - 6; http://dx.doi.org/10.1073/pnas.2032621100; PMID: 14504383
  • Richardson TE, Yu AE, Wen Y, Yang SH, Simpkins JW. Estrogen prevents oxidative damage to the mitochondria in Friedreich’s ataxia skin fibroblasts. PLoS One 2012; 7:e34600; http://dx.doi.org/10.1371/journal.pone.0034600; PMID: 22509330
  • Chang KCN, Wang Y, Oh IG, Jenkins S, Freedman LP, Thompson CC, et al. Estrogen receptor β is a novel therapeutic target for photoaging. Mol Pharmacol 2010; 77:744 - 50; http://dx.doi.org/10.1124/mol.109.062877; PMID: 20110405
  • Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, et al, Writing Group for the Women’s Health Initiative Investigators. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women’s Health Initiative randomized controlled trial. JAMA 2002; 288:321 - 33; http://dx.doi.org/10.1001/jama.288.3.321; PMID: 12117397
  • Jordan VC. Selective estrogen receptor modulation: a personal perspective. Cancer Res 2001; 61:5683 - 7; PMID: 11479197
  • Surazynski A, Jarzabek K, Haczynski J, Laudanski P, Palka J, Wolczynski S. Differential effects of estradiol and raloxifene on collagen biosynthesis in cultured human skin fibroblasts. Int J Mol Med 2003; 12:803 - 9; PMID: 14533013
  • Sumino H, Ichikawa S, Kasama S, Takahashi T, Kumakura H, Takayama Y, et al. Effects of raloxifene and hormone replacement therapy on forearm skin elasticity in postmenopausal women. Maturitas 2009; 62:53 - 7; http://dx.doi.org/10.1016/j.maturitas.2008.10.005; PMID: 19042101
  • Tham DM, Gardner CD, Haskell WL. Clinical review 97: Potential health benefits of dietary phytoestrogens: a review of the clinical, epidemiological, and mechanistic evidence. J Clin Endocrinol Metab 1998; 83:2223 - 35; http://dx.doi.org/10.1210/jc.83.7.2223; PMID: 9661587
  • Xu L, Lu A, Zhao X, Chen X, Cummings SR. Very low rates of hip fracture in Beijing, People’s Republic of China the Beijing Osteoporosis Project. Am J Epidemiol 1996; 144:901 - 7; http://dx.doi.org/10.1093/oxfordjournals.aje.a009024; PMID: 8890668
  • Hwang CS, Kwak HS, Lim HJ, Lee SH, Kang YS, Choe TB, et al. Isoflavone metabolites and their in vitro dual functions: they can act as an estrogenic agonist or antagonist depending on the estrogen concentration. J Steroid Biochem Mol Biol 2006; 101:246 - 53; http://dx.doi.org/10.1016/j.jsbmb.2006.06.020; PMID: 16965913
  • Muthyala RS, Ju YH, Sheng S, Williams LD, Doerge DR, Katzenellenbogen BS, et al. Equol, a natural estrogenic metabolite from soy isoflavones: convenient preparation and resolution of R- and S-equols and their differing binding and biological activity through estrogen receptors alpha and beta. Bioorg Med Chem 2004; 12:1559 - 67; http://dx.doi.org/10.1016/j.bmc.2003.11.035; PMID: 15018930
  • Jackson RL, Greiwe JS, Schwen RJ. Ageing skin: oestrogen receptor β agonists offer an approach to change the outcome. Exp Dermatol 2011; 20:879 - 82; http://dx.doi.org/10.1111/j.1600-0625.2011.01362.x; PMID: 21913999
  • Wang YN, Wu W, Chen HC, Fang H. Genistein protects against UVB-induced senescence-like characteristics in human dermal fibroblast by p66Shc down-regulation. J Dermatol Sci 2010; 58:19 - 27; http://dx.doi.org/10.1016/j.jdermsci.2010.02.002; PMID: 20211546
  • Robb EL, Stuart JA. Resveratrol interacts with estrogen receptor-β to inhibit cell replicative growth and enhance stress resistance by upregulating mitochondrial superoxide dismutase. Free Radic Biol Med 2011; 50:821 - 31; http://dx.doi.org/10.1016/j.freeradbiomed.2010.12.038; PMID: 21215799