ArticlePDF AvailableLiterature Review

The role of estrogen deficiency in skin ageing and wound healing

Authors:

Abstract and Figures

The links between hormonal signalling and lifespan have been well documented in a range of model organisms. For example, in C. elegans or D. melanogaster, lifespan can be modulated by ablating germline cells, or manipulating reproductive history or pregnenolone signalling. In mammalian systems, however, hormonal contribution to longevity is less well understood. With increasing age human steroid hormone profiles change substantially, particularly following menopause in women. This article reviews recent links between steroid sex hormones and ageing, with special emphasis on the skin and wound repair. Estrogen, which substantially decreases with advancing age in both males and females, protects against multiple aspects of cellular ageing in rodent models, including oxidative damage, telomere shortening and cellular senescence. Estrogen's effects are particularly pronounced in the skin where cutaneous changes post-menopause are well documented, and can be partially reversed by classical Hormone Replacement Therapy (HRT). Our research shows that while chronological ageing has clear effects on skin wound healing, falling estrogen levels are the principle mediator of these effects. Thus, both HRT and topical estrogen replacement substantially accelerate healing in elderly humans, but are associated with unwanted deleterious effects, particularly cancer promotion. In fact, much current research effort is being invested in exploring the therapeutic potential of estrogen signalling manipulation to reverse age-associated pathology in peripheral tissues. In the case of the skin the differential targeting of estrogen receptors to promote healing in aged subjects is a real therapeutic possibility.
Content may be subject to copyright.
REVIEW ARTICLE
The role of estrogen deficiency in skin ageing and wound
healing
Elaine Emmerson Matthew J. Hardman
Received: 26 November 2010 / Accepted: 11 February 2011
ÓSpringer Science+Business Media B.V. 2011
Abstract The links between hormonal signalling
and lifespan have been well documented in a range of
model organisms. For example, in C. elegans or
D. melanogaster, lifespan can be modulated by
ablating germline cells, or manipulating reproductive
history or pregnenolone signalling. In mammalian
systems, however, hormonal contribution to longev-
ity is less well understood. With increasing age
human steroid hormone profiles change substantially,
particularly following menopause in women. This
article reviews recent links between steroid sex
hormones and ageing, with special emphasis on the
skin and wound repair. Estrogen, which substantially
decreases with advancing age in both males and
females, protects against multiple aspects of cellular
ageing in rodent models, including oxidative damage,
telomere shortening and cellular senescence. Estro-
gen’s effects are particularly pronounced in the skin
where cutaneous changes post-menopause are well
documented, and can be partially reversed by clas-
sical Hormone Replacement Therapy (HRT). Our
research shows that while chronological ageing has
clear effects on skin wound healing, falling estrogen
levels are the principle mediator of these effects.
Thus, both HRT and topical estrogen replacement
substantially accelerate healing in elderly humans,
but are associated with unwanted deleterious effects,
particularly cancer promotion. In fact, much current
research effort is being invested in exploring the
therapeutic potential of estrogen signalling manipu-
lation to reverse age-associated pathology in periph-
eral tissues. In the case of the skin the differential
targeting of estrogen receptors to promote healing in
aged subjects is a real therapeutic possibility.
Keywords Estrogen Ageing Skin Wound
healing
Evolutionary conservation
The links between hormones and ageing, principally
identified through the study of model organisms, are
numerous. The most prominent example would be the
correlation between lifespan and insulin signalling,
which is conserved across numerous species. Indeed,
the importance of endocrine signalling in lower
animal species was initially demonstrated for nema-
tode (Caenorhabditis elegans) DAF-2, a homologue
of the mammalian insulin and insulin-like growth
factor (IGF) receptors, mutations in which lengthened
lifespan (Kimura et al. 1997; Kenyon et al. 1993;
Larsen et al. 1995). Mutations in numerous genes in
the insulin and IGF signalling pathway have also
been shown to prolong the lifespan of Drosophila
melanogaster (Tatar et al. 2001). Mice and higher
E. Emmerson M. J. Hardman (&)
Faculty of Life Sciences, The University of Manchester,
A V Hill Building, Oxford Road,
Manchester M13 9PT, UK
e-mail: matthew.j.hardman@manchester.ac.uk
123
Biogerontology
DOI 10.1007/s10522-011-9322-y
vertebrates, unlike worms and flies, have separate
insulin and IGF receptors. Interestingly, mice hetero-
zygous for IGF-1 receptor live *30% longer than
wild-type mice (Holzenberger et al. 2003), while
mice null for the downstream mediators, insulin
receptor substrate (IRS)-1 and -2, also have extended
lifespan (Selman et al. 2008; Taguchi et al. 2007).
Such extensive cross-species conservation confirms
the importance of this pathway in regulating lifespan
(Liang et al. 2003). In fact, decreasing insulin/IGF
signalling remains one of the few interventions that
does robustly extend murine longevity.
Reproductive state and reproductive system sig-
nals can also influence lifespan. For example, germ-
line precursor cell ablation in C. elegans and
D. melanogaster extends lifespan by *60% (Hsin
and Kenyon 1999; Flatt et al. 2008; Arantes-Oliveira
et al. 2002), a process thought to be regulated via
DAF-2 (Hsin and Kenyon 1999), DAF-9 [a homo-
logue of mammalian cytochrome P450] (Gerisch
et al. 2001; Gerisch and Antebi 2004; Jia et al. 2002)
and DAF-12 (Broue et al. 2007), amongst others
(Fig. 1a). The hormone repertoire of model organ-
isms most commonly used for ageing research, such
as C. elegans and D. melanogaster, is limited, yet
they do express hormone precursors present in higher
animals, such as pregnenolone and essential enzymes
necessary for conversion, such as cytochrome P450
(aromatase) (Broue et al. 2007; Motola et al. 2006).
Signalling via such precursors extends lifespan in
C. elegans (Yamawaki et al. 2010; Broue et al.
2007; Mak and Ruvkun 2004) and D. melanogaster
(Yamawaki et al. 2010; Simon et al. 2003). For
example, pregnenolone extends lifespan in nema-
todes, via DAF-9 (Broue et al. 2007) (Fig. 1b).
Surprisingly, the relevance of steroid precursors, such
as pregnenolone, in mammalian longevity is largely
unknown. Interestingly, aged ovariectomised (Ovx)
mice transplanted with the ovaries of young mice
exhibit significantly increased life expectancy
(Mason et al. 2009; Cargill et al. 2003), an effect
potentially mediated through the estrogen receptors
and/or the IGF-1R (Fig. 1c).
Ageing in humans, the menopause and pathology
With increasing age the human endocrine system
undergoes substantial change, particularly with
respect to hormones of adrenal origin. The steroid
precursor dehydroepiandrosterone (DHEA), its sul-
phate, DHEA-S, and the precursor androstendione
decline substantially in both males and females from
age 20 (Labrie et al. 1997). Interestingly, serum
concentrations of pregnenolone (discussed above)
also decrease with age in both males and females
(Havlikova et al. 2002; Labrie et al. 1997). In women
levels of sex steroid estrogens begin to fall from
approximately 35 years of age and follicle stimulat-
ing hormone (FSH) production is increased in an
effort to stimulate ovarian function (Chakravarti et al.
1976). While 17b-estradiol, the most potent form of
estrogen, decreases with age serum estrone concen-
trations remain fairly constant (Labrie et al. 1997).
The most pronounced changes occur when women
enter the menopause, a permanent cessation of
menstruation resulting from the loss of ovarian
follicular activity, which occurs at an average age
of 51 years in the developed world (Stanford et al.
Fig. 1 Association between steroid sex hormones and life-
span. aGermline cell ablation in C. elegans and D. melano-
gaster extends lifespan via DAF-2, -9 and -12. bPregnenolone
signals via DAF-9 and extends lifespan in C. elegans.
cHormone transfer by ovary transplant from young mice to
aged mice extends lifespan, an effect presumably mediated by
estrogen and the ERs or IGF-1R
Biogerontology
123
1987). Post-menopause the majority of circulating
sex steroids actually originate from circulating adr-
enally-derived DHEA (Labrie et al. 2011). In aged
males estrogen levels also decrease, although not as
rapidly as in females. By contrast, testosterone and
dihydrotestosterone (DHT) fluctuate but remain fairly
constant with age in both sexes (Labrie et al. 1997).
Of note, the enzyme 11b-hydroxysteroid hydrogenase
(HSD), which is responsible for the conversion of
cortisone to cortisol, is markedly increased with age
(Vukelic et al. 2011; Tiganescu et al. 2011).
Increasing life expectancy now means that the
average woman in the developed world spends one-
third of her life in the post-menopausal period
(Kligman and Koblenzer 1997). It is widely accepted
that decreased systemic hormones in the post-men-
opausal state is associated with increased risk for a
range of age-associated pathologies (Table 1).
Estrogen deficiency as a general mechanism
of ageing
Arguably the most profound hormonal change in
ageing is the post-menopause reduction in 17b-
estradiol. Moreover, a substantial body of literature
links estrogen decline to the distinct cellular ageing
mechanisms of oxidative damage and cellular
senescence.
Oxidative damage
Free radicals are strongly implicated in the cellular
damage that accompanies ageing and age-associated
disease (Harman 1956; Beckman and Ames 1998),
part of the ‘mitochondrial theory of ageing’ (Miquel
et al. 1980). The link between mitochondria and
longevity comes from the observation that by reduc-
ing mitochondrial peroxidase production lifespan can
be extended (Lopez-Torres et al. 1993; Ku et al.
1993). Pertinent to this review is the observation that
both peroxidase production and mitochondrial DNA
damage are significantly (40–80%) higher in male
rats than in age-matched females, and this is directly
attributed to differences in estrogen levels (Borras
et al. 2003; Pinto and Bartley 1968,1969). Mito-
chondrial glutathione, a biological marker of ageing
and age-associated damage (Hazelton and Lang 1984;
Sastre et al. 2000) is significantly higher in male rats
than females (Vina et al. 2005). Conversely, expres-
sion of 16S ribosomal RNA, a biological marker of
youth (Calleja et al. 1993), is substantially higher in
female rats than males (Borras et al. 2003; Vina et al.
2005). Although, Sanz et al. (2007) report no
difference in oxidative stress, age-related damage or
lifespan between male and female C57Bl6 mice, Ali
et al. (2006) show that females, the reported shorter
lived gender, exhibit enhanced reactive oxygen
species (ROS) production with age.
Table 1 Chronic pathologies
associated with ageing and
menopause
Pathology Age Menopause
Auditory degeneration Weinstein and Ventry (1982) Hederstierna et al. (2010)
Hederstierna et al. (2007)
Cancer Yancik et al. (2001) Trichopoulos et al. (1972)
Heart disease Lindblad et al. (2001) Lokkegaard et al. (2006)
Lye and Donnellan (2000) Jacobsen et al. (1997)
Colditz et al. (1987) Barrett-Connor (1995)
Muscular degeneration Evans (2010) Calmels et al. (1995)
Neurodegeneration Kukull et al. (2002) Simpkins et al. (2005)
Suthers et al. (2003) Morrison et al. (2006)
Optical degeneration Owsley et al. (2007) Siesky et al. (2008)
Carcenac et al. (2009) Evans et al. (1998)
Osteoporosis Gates et al. (2009) Melton et al. (1992)
Simonen and Mikkola (1991)
Skin ageing Lavker (1979) Brincat et al. (1987)
Gilchrest et al. (1983) Sumino et al. (2004)
Urinary incontinence Williams and Pannill (1982) Waetjen et al. (2009)
Biogerontology
123
Estrogen deficiency post-menopause is also
strongly linked to altered oxidative state, with
estrogen a potent direct antioxidant and indirect
inducer of antioxidant enzymes. In ovariectomised
(Ovx) female rats oxidised glutathione, lipid perox-
idation and mitochondrial DNA damage are signif-
icantly increased, and can be reversed by estrogen
replacement or phytoestrogen treatment (Baeza et al.
2010). In vitro both estradiol and the phytoestrogen
genistein reduce the production of hydrogen peroxide
in MCF-7 cells (Borras et al. 2005,2006). Addition-
ally, keratinocytes isolated from aged female rats
exhibit increased oxidative stress (lipoperoxides) and
apoptosis (caspases 3 and 8), which can be reversed
by estrogen treatment (Tresguerres et al. 2008).
Finally, it should be noted that while gender-specific
effects are common across many species, including
man (Gurwitz 2005), marsupials (Humphries and
Stevens 2001) and non-human primates (Herndon
et al. 1999) this is not the case in all animal species.
Some species lack gender difference in lifespan (Sanz
et al. 2007; Wich et al. 2004) while in a limited
number of species males actually live longer (Asdell
and Joshi 1976; McCulloch and Gems 2003).
Senescence
Significant telomere shortening is associated with
replicative cell senescence and tissue deterioration
(Allsopp et al. 1992; Harley 1991), and prevented by
the enzyme telomerase (Morin 1989). The expression
of the human telomerase reverse transcriptase
(hTERT) gene and telomerase activity (TA) is
upregulated in hepatocytes in vitro in response to
estradiol, leading to maintained telomere length (Sato
et al. 2004). Similarly, estrogen dose-dependently
prevents cellular senescence and increases telomerase
activity in endothelial progenitor cells (Imanishi et al.
2005b), vascular smooth muscle cells (Ling et al.
2006) and leukocytes (Aviv et al. 2006) in vitro.In
endothelial progenitor cells estrogen prevents angio-
tensin-II-mediated oxidative stress and senescence
(Imanishi et al. 2005a). Estrogen deficiency signifi-
cantly inhibits TERT expression and telomerase
activity in the mouse adrenal gland, an effect
reversed by estrogen replacement (Bayne et al.
2008). It has thus been speculated that gender specific
differences in longevity are, at least in part, due to
hormonal regulation of telomere function (Aviv et al.
2005; Stindl 2004). One method of detecting senes-
cent cells in vitro is a modified beta-galactosidase (b-
gal) assay (Dimri et al. 1995). Interestingly, the soy-
derived phytoestrogen genistein suppresses UVB-
induced expression of b-gal in primary human dermal
fibroblasts (Wang et al. 2010), while soybean extract
protects against cellular senescence in HaCaT cells
(Chiu et al. 2009).
Skin ageing and estrogens
With increasing age a combination of intrinsic and
extrinsic factors (primarily UV exposure) lead to skin
deterioration. Aged skin has altered structure and
reduced function, particularly loss of elasticity,
wrinkling, thinning and fragility. While there are
differences in the etiology of intrinsic and extrinsic
ageing (Tsoureli-Nikita et al. 2006) the majority of
resultant changes are similar (Tsoureli-Nikita et al.
2006; Pillai et al. 2005; Ashcroft et al. 1997e; Varani
et al. 2001; Lavker 1979). Specifically, intrinsic
ageing involves thinning of the epidermis and dermis,
reduced epidermal proliferation and turnover and
reduced vascularity (Gilchrest et al. 1982b; Lavker
1979). Collagen production is reduced (Shuster et al.
1975) and distribution is altered (Richard et al. 1993),
while the production of matrix degrading enzymes
(MMPs) is increased with intrinsic ageing (Ashcroft
et al. 1997e). Extrinsic ageing or photoageing
involves dermal elastosis (Mitchell 1967; Braverman
and Fonferko 1982), reduced Langerhans cell num-
bers, altered melanocyte distribution (Thiers et al.
1984; Gilchrest et al. 1982a) and increased MMP
activity (Pillai et al. 2005). With advancing age
reduced sebaceous gland secretion leads to skin
xerosis, an event that involves corticotropin-releasing
hormone (CRH) and coincides with the onset of
menopause in women (Pochi et al. 1979; Zouboulis
et al. 2002).
The effects of ageing can be readily observed in
skin cells in vitro, where dermal fibroblasts and
epidermal keratinocytes from aged donors have
reduced proliferative capacity and display premature
senescence (Stanulis-Praeger and Gilchrest 1989;
Gilchrest 1983; Schneider and Mitsui 1976; Mets
et al. 1983). In ageing human skin senescent cells
accumulate through a combination of growth arrest
and increased resistance to apoptosis (Dimri et al.
Biogerontology
123
1994; Wang 1995). In the principal skin cell types
(keratinocytes/fibroblasts) telomeres shorten an aver-
age of 9 and 11 base pairs per year, respectively,
throughout life (Sugimoto et al. 2006; Krunic
et al. 2009). Interestingly, telomere shortening is
reportedly unchanged in sun-exposed (extrinsically
aged) versus sun-protected (intrinsically aged) skin
(Sugimoto et al. 2006). Instead, large deletions of the
mitochondrial genome are believed to be involved in
UV-induced skin photoageing (Schroeder et al.
2008). In vitro, repetitive exposure of human dermal
fibroblasts to UVA has been shown to induce
deletions of up to 5000 base pairs, causing a partial
loss of the mitochondrial genome (Berneburg et al.
1999). Moreover, in a model where mtDNA deletions
were induced without irradiation, gene expression
mirrored that observed in photoaged skin (Schroeder
et al. 2008).
While laboratory animals provide excellent trac-
table models, some aspects of skin ageing, e.g.,
changes in epidermal thickness, are contentious and
appear species and strain dependent. The majority of
mouse studies find that with age epidermal thickness
is reduced (Iversen and Schjoelberg 1984; Haratake
et al. 1997; Bhattacharyya and Thomas 2004;
Argyris 1983) and epidermal keratinocyte prolifer-
ation is decreased (Cameron 1972). However, other
studies have reported increased epidermal thickness
with age (Hill 1988). Epidermal thickness in aged
rats has been reported to increase (Bhattacharyya
et al. 2005; Thomas 2005), decrease (Morris et al.
1990) or remain unchanged (Giangreco et al. 2008;
Monteiro-Riviere et al. 1991). Interestingly, Ishib-
ashi rats (progeny of Wistar and wild rats) report-
edly undergo qualitatively similar skin changes to
humans. From 12 weeks of age wrinkling of the
skin occurs, consistent with a reduction in elastin
(Sakuraoka et al. 1996). Calorie restriction (CR),
which extends lifespan in numerous model organ-
isms (S. cerevisiae,C. elegans,D. melanogaster)
(Lin et al. 2002; Klass 1977; Hosono et al. 1989;
Mair et al. 2003; Pletcher et al. 2002) and higher
mammals, such as mice and non-human primates
(Lane et al. 1995; McCay et al. 1935; Rezzi et al.
2009; Ramsey et al. 2000), prevents age-related skin
changes when compared to age-matched ad libitum
controls (Thomas 2005; Bhattacharyya et al. 2005).
Unfortunately, neither of these studies specifies the
sex of the experimental animals.
Historic studies provide the first evidence of
estrogen’s cutaneous effects: topically applied follic-
ular hormone, now known to be estrogen, was found
to locally improve acne and eczema (Loeser 1937). It
has also long been known that the symptoms of skin
disorders such as psoriasis improve during preg-
nancy, an observation now directly attributed to
increased circulating estrogen (Dunna and Finlay
1989; Boyd et al. 1996) Moreover, the oral contra-
ceptive pill is often prescribed to treat severe acne
(Arowojolu et al. 2009). During the menopause, skin
undergoes major changes which include reduced
epidermal and dermal thickness, a decrease in
collagen content (Brincat et al. 1987), reduced
elasticity (Sumino et al. 2004), dryness and fragility
(Brincat et al. 1985). Crucially, the majority of these
changes can be reversed by either topical or systemic
hormone replacement therapy (HRT) (Table 2). Not
surprisingly, topical estrogen treatment has little
effect on young skin (Goldzieher 1949). The effects
of estrogen are predominantly mediated through the
estrogen receptors (see ‘Estrogen receptors and
SERMs’ section). Estrogen also protects against
photoageing, with mortality rates from both mela-
noma (Miller and Mac Neil 1997) and non-melanoma
(Weinstock 1994) skin cancer lower in women than
men. In experimental animal studies estrogen depri-
vation enhances sensitivity to UV skin damage and
accelerates photoageing, measured as wrinkling, a
loss of elasticity and damage to elastic fibres
(Tsukahara et al. 2001; Tsukahara et al. 2004), while
estrogen treatment increases skin collagen content in
rats and guinea pigs (Smith and Allison 1966;
Henneman 1968), increases hyaluronic acid synthesis
(Sobel et al. 1965) and promotes epidermal thicken-
ing in mice (Bullough 1947). A potential link
between ageing and estrogen involves IGF-1. Estra-
diol is known to be able to signal through the IGF-1R
in the skin, in a non-genomic manner, strongly
implicating it’s involvement in the regulation of
cutaneous ageing (Makrantonaki et al. 2008;
Surazynski et al. 2003).
In reality, the skin is not only an estrogen target,
but also a major synthetic organ with the capacity to
release and produce a wide range of hormones,
including estrogens. Skin cells are able to synthesise
locally acting estrogens from the precursors choles-
terol and DHEA (Fig. 2) (Simpson et al. 1997; Bulun
et al. 1999; Chen et al. 2002; Chen et al. 1998;
Biogerontology
123
Thiboutot et al. 1998; Hughes et al. 1997; Sawaya
and Penneys 1992; Dumont et al. 1992; Eicheler et al.
1995; Thiboutot et al. 2000). Additionally, local
production of 11b-HSD by epidermal keratinocytes,
dermal fibroblasts and hair follicles (Tiganescu et al.
2011) increases cortisol conversion and has been
speculated to control the inflammatory response
(Vukelic et al. 2011). In aged individuals peripheral
hormone production and local hormone signalling
are likely to be particularly important, in light
of the considerable reduction in systemic hormone
production (Longcope 1971). Surprisingly, the effects
of ageing on peripheral hormone synthesis/conver-
sion are virtually unknown.
Wound repair with age
Cutaneous wound healing is a complex tightly
orchestrated response to injury, carefully regulated
at temporal and spatial levels (reviewed in Shaw and
Martin 2009). In young individuals the innate
Table 2 Cutaneous changes
with post-menopausal
estrogen replacement
Cutaneous changes Reference(s)
Increased epidermal thickness Punnonen (1971), Brincat (2000), Fuchs et al. (2003)
Increased dermal thickness Son et al. (2005), Shuster et al. (1975), Brincat et al. (1985)
Increased collagen content Castelo-Branco et al. (1992), Savvas et al. (1993), Shuster
et al. (1975), Varila et al. (1995)
Increased elastin content Punnonen et al. (1987)
Increased skin moisture content Pierard-Franchimont et al. (1995), Schmidt et al. (1994)
Improved external appearance Schmidt et al. (1996), Schmidt et al. (1994), Brincat et al.
(1985), Creidi et al. (1994)
Fewer wrinkles Aertgeerts (1974)
Fig. 2 Skin contains all the components for peripheral
steroid hormone synthesis. The precursors cholesterol and
dehydroepiandrosterone (DHEA) can be converted to steroid
hormones by aromatase (the product of the CYP19 gene), 3b-
hydroxysteroid dehydrogenase (3b-HSD), 17b-hydroxysteroid
dehydrogenase (17b-HSD) and 5a-reductase. 17a-estradiol has
been shown to influence the conversion of testosterone to 17b-
estradiol and androstenedione. Double boxes represent func-
tionally important non-steroid hormone synthesis
Biogerontology
123
inflammatory response is first initiated, followed by a
widespread proliferative phase, involving fibroblasts,
keratinocytes and endothelial cells. Keratinocyte
migration restores the skin’s barrier, fibroblast-med-
iated contraction aids wound closure, while matrix
remodelling leads to a mature scar. With the passage
of time skin becomes fragile and prone to trauma,
while cellular ageing leads to aberrant healing.
Platelet function (Boldt et al. 2001; Fukaya et al.
2000) and platelet-derived growth factor (PDGF)
expression (Ashcroft et al. 1997d) is altered with age.
The inflammatory response becomes disrupted, with
excessive neutrophil influx, altered endothelial cell
adhesion, prolonged macrophage recruitment and
delayed resolution (Ashcroft et al. 1998). This leads
to over-production of matrix degrading enzymes,
classically elastase and MMPs (Ashcroft et al. 1997e;
Ashcroft et al. 1997f; Herrick et al. 1997), and under-
expression of tissue inhibitors of metalloproteinases
(TIMPs) (Ashcroft et al. 1997b). In vivo, re-epithel-
ialisation is delayed in aged humans and murine
models (Ashcroft et al. 1997a). Corroborative in vitro
studies show that keratinocytes from young donors
proliferate at a faster rate (Stanulis-Praeger and
Gilchrest 1986) and exhibit less sensitivity to epider-
mal growth factor (EGF) and keratinocyte growth
factor (KGF) (Gilchrest 1983) than those from old
aged donors. Angiogenesis is delayed in aged humans
and mice (Ashcroft et al. 1997c; Sadoun and Reed
2003) and collagen deposition is reduced (Lenhardt
et al. 2000; Ashcroft et al. 1997c). Ultimately, in
elderly subjects scar strength is reduced (Lindstedt
and Sandblom 1975; Sandblom et al. 1953; Mendoza
et al. 1970), however, scar quality is improved
(Ashcroft et al. 1999).
Repair, ageing and hormones
While chronological age is a clear risk factor for poor
healing our recent studies suggest that estrogen
deprivation is the major factor controlling delayed
healing in elderly humans. In our recent microarray
study 78% of genes differentially expressed between
wounds from young and elderly men were estrogen-
regulated, while only 3% were age-associated,
strongly implicating reduced estrogen, and not known
gerontogenes, as the primary regulator of delayed
healing in aged subjects (Hardman and Ashcroft
2008). In post-menopausal women HRT improves
healing (Ashcroft et al. 1997a), while topical estrogen
treatment improves healing in elderly subjects of
either gender (Ashcroft et al. 1999). Estrogen’s
cellular effects include dampening excessive neutro-
phil recruitment, preventing disproportionate elastase
production, and increasing fibronectin and collagen
deposition. Crucially, two independent studies reveal
that HRT protects post-menopausal women from
developing venous leg ulcers or pressure ulcers
(Margolis et al. 2002; Berard et al. 2001).
Animal models have provided key insight into
estrogen’s role in wound healing (summarised in
Fig. 3). Estrogen replacement accelerates cutaneous
healing in Ovx female mice and rats (Ashcroft et al.
Fig. 3 Mechanism of
estrogen activity in older
people. Estrogen aids
healing through effects on
fibroblasts, keratinocytes
and inflammatory cells
Biogerontology
123
1997a; Ashcroft et al. 2003; Emmerson et al. 2010;
Hardman et al. 2008) and male rats (Rajabi and
Rajabi 2007). Systemic treatment with the sex steroid
precursor DHEA accelerates wound healing in young
Ovx female mice and old male mice, a result
attributed to the local conversion of DHEA to
estrogen (Mills et al. 2005). Of interest, androgens
are detrimental to healing. The testosterone metabo-
lite DHT retards repair (Gilliver et al. 2009), while
castration or androgen receptor blockade improves
healing in rodents (Gilliver et al. 2003; Gilliver et al.
2006; Ashcroft and Mills 2002).
At the cellular level estrogen down-regulates
neutrophil-expressed L-selectin, preventing the
excessive neutrophil accumulation and neutrophil-
derived elastase production characteristic of aged
healing (Ashcroft et al. 1999), and improves neutro-
phil phagocytic ability (Magnusson and Einarsson
1990). Estrogen dampens expression of numerous
pro-inflammatory cytokines, including MIF, TNFa,
MCP-1, IL-1band IL-6 (Kovacs et al. 1996; Hu et al.
1988). The reduction in Macrophage Migration
Inhibitory Factor (MIF), in particular, plays a major
role in the beneficial effects of estrogen on wound
repair (Ashcroft et al. 2003; Hardman et al. 2005;
Emmerson et al. 2009). Of interest, plasma MIF
levels, which inversely correlate with systemic
estrogen (Aloisi et al. 2005), are increased post-
menopause and fall following HRT (Hardman et al.
2005). Estrogen is also a keratinocyte mitogen
(Verdier-Sevrain et al. 2004), promoting migration
across an artificial scratch in vitro (Emmerson et al.
2009; Campbell et al. 2010) and wound re-epithel-
ialisation in Ovx mice (Emmerson et al. 2010;
Hardman et al. 2008). Moreover, impaired wound
re-epithelialisation post-menopause can be reversed
to pre-menopause levels by just 3 months of HRT
(Ashcroft et al. 1997a). Estrogen directly stimulates
dermal fibroblast migration in vitro (Emmerson et al.
2009; Campbell et al. 2010) and indirectly via
stimulating macrophage platelet derived growth fac-
tor (PDGF), a key fibroblast mitogen and stimulator
of wound contraction (Battegay et al. 1994). The role
of estradiol in stimulating angiogenesis is somewhat
contentious. PDGF directly stimulates angiogenesis,
while estradiol increases endothelial cell capillary-
like structure formation upon reconstituted basement
membrane (Morales et al. 1995). However, conflict-
ing in vitro studies report either no change or
decreased angiogenesis following estrogen treatment
(Nyman 1971; Lundgren 1973).
Estrogen receptors and SERMs
Estrogen signals via two nuclear hormone receptors,
ERaand ERb.ERa, the first receptor to be identified
and cloned (Walter et al. 1985), predominates in
reproductive tissues and is strongly associated with
cancer (Zou and Ing 1998; Kuiper et al. 1997; Ali and
Coombes 2000). The second receptor, ERb, identified
over a decade later (Kuiper et al. 1996; Ogawa et al.
1998), is more highly expressed in peripheral, non-
reproductive tissues (Kuiper et al. 1997; Onoe et al.
1997; Brandenberger et al. 1997; Couse et al. 1997).
Both receptors are reportedly expressed in human
facial skin (Miller and Mac Neil 1997), scalp (Thorn-
ton et al. 2003a,b; Mosselman et al. 1996) and upper
arm skin (Reed et al. 2005). Several studies suggest that
ERbpredominates in keratinocytes of human scalp
skin (Mosselman et al. 1996; Thornton et al. 2003a,b)
while others identify both receptors in neonatal
foreskin-derived keratinocytes (Verdier-Sevrain et al.
2004). In mouse skin both receptors are widely
expressed (Campbell et al. 2010; Cho et al. 2008).
Our recent data indicate that delayed healing in
Ovx female mice can be reversed by stimulating
signalling through ERbalone (using the ERb-specific
agonist DiarylPropioNitrile [DPN]) (Campbell et al.
2010). Conversely, signalling through ERaalone
(using the ERaagonist Propyl Pyrazole Triol [PPT])
entirely fails to improve healing in Ovx mice. To
support this finding, we find that estrogen replace-
ment in Ovx mice lacking functional ERb(ERb-/-)
actually further delays healing beyond the Ovx wild-
type phenotype. Moreover, epidermal specific ERb
null mice (K14-cre/ERb
L2/L2
) phenocopy ERb-/-
mice, i.e., estrogen replacement again impairs heal-
ing. This would suggest that the beneficial effects of
estrogen on cutaneous repair are predominantly
mediated via epidermal ERb(Campbell et al.
2010). Of note, the beneficial effects of 17b-estradiol
on outcome in a skin flap necrosis model is reportedly
mediated via ERa(Toutain et al. 2009) while Ovx
rats treated with PPT exhibit reduced wound tensile
strength (Gal et al. 2010). A crucial link between our
mouse data and aged human healing is provided by
the observation that polymorphisms in the human
Biogerontology
123
ERbgene are significantly associated with venous
ulceration in the Caucasian population (Ashworth
et al. 2005,2008).
Most pathological states in the elderly involve a
single ER isoform, for example in cancers of the
reproductive system ERapredominates (Herynk and
Fuqua 2004; Yang et al. 2008; Cai et al. 2003), while
in colon cancers ERbpredominates (Arai et al. 2000;
Fiorelli et al. 1999; Foley et al. 2000; Qiu et al. 2002).
The clear differential roles of the two estrogen
receptors in cutaneous repair (Campbell et al. 2010;
Gal et al. 2010; Toutain et al. 2009) suggest that
pharmacological manipulation may be a viable ther-
apeutic option. Compounds termed Selective Estro-
gen Receptor Modulator (SERMs) are being
developed to treat pathologies such as osteoporosis
and breast cancer by exploiting natural estrogen
signalling to confer tissue specific estrogenic or anti-
estrogenic effects.
SERMs have been developed to provide estrogen-
like beneficial effects, to treat disorders such as
osteoporosis, an approach that should bypass
systemic estrogen risks, including breast cancer
(Matsumoto 2006). Arguably, the best characterised
and commonly used SERMs are tamoxifen and
raloxifene. Both are considered ER antagonists in
the breast, but have the potential to act as ER agonists
in other tissues (Frasor et al. 2004). The dietary
phytoestrogen genistein, is also becoming increas-
ingly popular for the treatment of post-menopausal
pathology (Atteritano et al. 2008; D’Anna et al.
2007). Despite use of SERMs in age- and menopause-
associated pathologies (reviewed in Pickar et al.
2010) knowledge of SERM activity in the skin is
severely lacking. In vitro both tamoxifen and raloxif-
ene stimulate fibroblast proliferation, but fail to
promote fibroblast migration (Stevenson et al.
2009). Preliminary studies in post-menopausal skin
indicate increased elasticity and collagen content
following raloxifene treatment (Sumino et al. 2009)
and improved dermal vascularisation and increased
epidermal thickness following genistein treatment
(Moraes et al. 2009). Topical tamoxifen reportedly
improves the appearance of keloid scars in acute
burns patients, through dampening of fibroblast
proliferation and collagen synthesis (Mousavi et al.
2010; Gragnani et al. 2009). Our recent studies
indicate that the SERMs tamoxifen, raloxifene and
genistein all substantially benefit healing in the
estrogen-deprived Ovx mouse, promoting re-epithel-
ialisation and contraction, and reducing inflamma-
tion, an effect that is most likely mediated via ERb
(Emmerson et al. 2010; Hardman et al. 2008), while a
separate study provides evidence that genistein
accelerates murine healing by modulating TGFb1
(Marini et al. 2010). Tamoxifen, raloxifene and
genistein are anti-inflammatory in other pathologies,
including systemic lupus erythematosus (SLE)
(Sthoeger et al. 2003), stroke (Tian et al. 2009),
UV-associated cutaneous damage (Brand and
Jendrzejewski 2008; Shyong et al. 2002; Widyarini
et al. 2001), colitis (Seibel et al. 2009), ileitis
(Sadowska-Krowicka et al. 1998) and multiple scle-
rosis (MS) (De Paula et al. 2008). An important and
on-going goal of our research is to evaluate cutaneous
healing in post-menopausal women prescribed topical
SERM treatment.
Conclusions and further perspectives
Despite links between estrogen and ageing being
suggested many years ago, only over the last decade
has estrogen emerged as a key determinant of ageing
in peripheral non-reproductive tissues, particularly
bone, skin and brain. Indeed, the endocrine theory of
ageing, underpinned by germline cell ablation exper-
iments in model organisms, states that chronological
changes in hormones levels accelerate the cellular
effects of ageing. In skin particularly, it is only over
the last few years that we have begun to understand
the relative contributions of hormones and ageing to
pathological healing. Indeed, although our knowledge
of estrogen’s beneficial effects on wound repair has
greatly expanded over recent years there is still much
that is not understood. The prospect of manipulating
estrogen signalling to alleviate poor healing in the
elderly is an exciting one, but we have yet to fully
understand how such ER-mediated signalling
changes with normal ageing and how this impacts
upon cutaneous pathology post-menopause. Impor-
tantly, how do the protective effects of estrogen on
cellular ageing (i.e., prevention of telomere shorten-
ing and oxidative stress) directly contribute to
improved healing? A clearer detailed mechanistic
understanding will be of relevance to a range of
peripheral estrogen-target tissues and aid the
Biogerontology
123
development of targeted hormone-based therapies, to
promote cutaneous repair in the elderly.
Acknowledgments Dr Emmerson is the David Hammond
Charitable Trust Research Associate (The Healing Foundation)
and recipient of the BSRA 2010 Korenchevsky award. Dr
Hardman is the Edmund de Rothschild Senior Fellow in
Ageing Research (Research Into Ageing; Age UK).
References
Aertgeerts J (1974) Effect of oral estrogens on the skin of the
woman in post-menopause or after castration. Ther Ggw
113(10):1739–1748
Ali S, Coombes RC (2000) Estrogen receptor alpha in human
breast cancer: occurrence and significance. J Mammary
Gland Biol Neoplasia 5(3):271–281
Ali SS, Xiong C, Lucero J, Behrens MM, Dugan LL, Quick KL
(2006) Gender differences in free radical homeostasis
during aging: shorter-lived female c57bl6 mice have
increased oxidative stress. Aging Cell 5(6):565–574
Allsopp RC, Vaziri H, Patterson C, Goldstein S, Younglai EV,
Futcher AB, Greider CW, Harley CB (1992) Telomere
length predicts replicative capacity of human fibroblasts.
Proc Natl Acad Sci USA 89(21):10114–10118
Aloisi AM, Pari G, Ceccarelli I, Vecchi I, Ietta F, Lodi L,
Paulesu L (2005) Gender-related effects of chronic non-
malignant pain and opioid therapy on plasma levels of
macrophage migration inhibitory factor (mif). Pain
115(1–2):142–151
Arai N, Strom A, Rafter JJ, Gustafsson JA (2000) Estrogen
receptor beta mRNA in colon cancer cells: growth effects
of estrogen and genistein. Biochem Biophys Res Commun
270(2):425–431
Arantes-Oliveira N, Apfeld J, Dillin A, Kenyon C (2002)
Regulation of life-span by germ-line stem cells in cae-
norhabditis elegans. Science 295(5554):502–505
Argyris TS (1983) The effect of aging on epidermal mass in
balb/c female mice. Mech Ageing Dev 22(3–4):347–354
Arowojolu AO, Gallo MF, Lopez LM, Grimes DA, Garner SE
(2009) Combined oral contraceptive pills for treatment of
acne. Cochrane Database Syst Rev (3):CD004425
Asdell SA, Joshi SR (1976) Reproduction and longevity in the
hamster and rat. Biol Reprod 14(4):478–480
Ashcroft GS, Mills SJ (2002) Androgen receptor-mediated
inhibition of cutaneous wound healing. J Clin Invest
110(5):615–624
Ashcroft GS, Dodsworth J, van Boxtel E, Tarnuzzer RW,
Horan MA, Schultz GS, Ferguson MW (1997a) Estrogen
accelerates cutaneous wound healing associated with an
increase in tgf-beta1 levels. Nat Med 3(11):1209–1215
Ashcroft GS, Herrick SE, Tarnuzzer RW, Horan MA, Schultz
GS, Ferguson MW (1997b) Human ageing impairs injury-
induced in vivo expression of tissue inhibitor of matrix
metalloproteinases (timp)-1 and -2 proteins and mrna.
J Pathol 183(2):169–176
Ashcroft GS, Horan MA, Ferguson MW (1997c) Aging is
associated with reduced deposition of specific
extracellular matrix components, an upregulation of
angiogenesis, and an altered inflammatory response in a
murine incisional wound healing model. J Invest Derma-
tol 108(4):430–437
Ashcroft GS, Horan MA, Ferguson MW (1997d) The effects of
ageing on wound healing: immunolocalisation of growth
factors and their receptors in a murine incisional model.
J Anat 190(Pt 3):351–365
Ashcroft GS, Horan MA, Herrick SE, Tarnuzzer RW, Schultz
GS, Ferguson MW (1997e) Age-related differences in the
temporal and spatial regulation of matrix metalloprotein-
ases (mmps) in normal skin and acute cutaneous wounds
of healthy humans. Cell Tissue Res 290(3):581–591
Ashcroft GS, Kielty CM, Horan MA, Ferguson MW (1997f)
Age-related changes in the temporal and spatial distribu-
tions of fibrillin and elastin mrnas and proteins in acute
cutaneous wounds of healthy humans. J Pathol
183(1):80–89
Ashcroft GS, Horan MA, Ferguson MW (1998) Aging alters
the inflammatory and endothelial cell adhesion molecule
profiles during human cutaneous wound healing. Lab
Invest 78(1):47–58
Ashcroft GS, Greenwell-Wild T, Horan MA, Wahl SM, Fer-
guson MW (1999) Topical estrogen accelerates cutaneous
wound healing in aged humans associated with an altered
inflammatory response. Am J Pathol 155(4):1137–1146
Ashcroft GS, Mills SJ, Lei K, Gibbons L, Jeong MJ, Taniguchi
M, Burow M, Horan MA, Wahl SM, Nakayama T (2003)
Estrogen modulates cutaneous wound healing by down-
regulating macrophage migration inhibitory factor. J Clin
Invest 111(9):1309–1318
Ashworth JJ, Smyth JV, Pendleton N, Horan M, Payton A,
Worthington J, Ollier WE, Ashcroft GS (2005) The
dinucleotide (ca) repeat polymorphism of estrogen
receptor beta but not the dinucleotide (ta) repeat poly-
morphism of estrogen receptor alpha is associated with
venous ulceration. J Steroid Biochem Mol Biol
97(3):266–270
Ashworth JJ, Smyth JV, Pendleton N, Horan M, Payton A,
Worthington J, Ollier WE, Ashcroft GS (2008) Poly-
morphisms spanning the on exon and promoter of the
estrogen receptor-beta (erbeta) gene esr2 are associated
with venous ulceration. Clin Genet 73(1):55–61
Atteritano M, Pernice F, Mazzaferro S, Mantuano S, Frisina A,
D’Anna R, Cannata ML, Bitto A, Squadrito F, Frisina N,
Buemi M (2008) Effects of phytoestrogen genistein on
cytogenetic biomarkers in postmenopausal women: 1 year
randomized, placebo-controlled study. Eur J Pharmacol
589(1–3):22–26
Aviv A, Shay J, Christensen K, Wright W (2005) The lon-
gevity gender gap: are telomeres the explanation? Sci
Aging Knowledge Environ 2005(23):pe16
Aviv A, Valdes A, Gardner JP, Swaminathan R, Kimura M,
Spector TD (2006) Menopause modifies the association of
leukocyte telomere length with insulin resistance and
inflammation. J Clin Endocrinol Metab 91(2):635–640
Baeza I, Fdez-Tresguerres J, Ariznavarreta C, De la Fuente M
(2010) Effects of growth hormone, melatonin, oestrogens
and phytoestrogens on the oxidized glutathione (gssg)/
reduced glutathione (gsh) ratio and lipid peroxidation in
aged ovariectomized rats. Biogerontology 11(6):687–701
Biogerontology
123
Barrett-Connor E (1995) Postmenopausal estrogen and heart
disease. Atherosclerosis 118(Suppl):S7–S10
Battegay EJ, Rupp J, Iruela-Arispe L, Sage EH, Pech M (1994)
Pdgf-bb modulates endothelial proliferation and angio-
genesis in vitro via pdgf beta-receptors. J Cell Biol
125(4):917–928
Bayne S, Jones ME, Li H, Pinto AR, Simpson ER, Liu JP
(2008) Estrogen deficiency leads to telomerase inhibition,
telomere shortening and reduced cell proliferation in the
adrenal gland of mice. Cell Res 18(11):1141–1150
Beckman KB, Ames BN (1998) The free radical theory of
aging matures. Physiol Rev 78(2):547–581
Berard A, Kahn SR, Abenhaim L (2001) Is hormone replace-
ment therapy protective for venous ulcer of the lower
limbs? Pharmacoepidemiol Drug Saf 10(3):245–251
Berneburg M, Grether-Beck S, Kurten V, Ruzicka T, Briviba
K, Sies H, Krutmann J (1999) Singlet oxygen mediates the
uva-induced generation of the photoaging-associated
mitochondrial common deletion. J Biol Chem 274(22):
15345–15349
Bhattacharyya TK, Thomas JR (2004) Histomorphologic
changes in aging skin: observations in the cba mouse
model. Arch Facial Plast Surg 6(1):21–25
Bhattacharyya TK, Merz M, Thomas JR (2005) Modulation of
cutaneous aging with calorie restriction in Fischer 344
rats: a histological study. Arch Facial Plast Surg
7(1):12–16
Boldt J, Huttner I, Suttner S, Kumle B, Piper SN, Berchthold G
(2001) Changes of haemostasis in patients undergoing
major abdominal surgery—is there a difference between
elderly and younger patients? Br J Anaesth 87(3):435–440
Borras C, Sastre J, Garcia-Sala D, Lloret A, Pallardo FV, Vina
J (2003) Mitochondria from females exhibit higher anti-
oxidant gene expression and lower oxidative damage than
males. Free Radic Biol Med 34(5):546–552
Borras C, Gambini J, Gomez-Cabrera MC, Sastre J, Pallardo
FV, Mann GE, Vina J (2005) 17beta-oestradiol up-regu-
lates longevity-related, antioxidant enzyme expression via
the erk1 and erk2[mapk]/nfkappab cascade. Aging Cell
4(3):113–118
Borras C, Gambini J, Gomez-Cabrera MC, Sastre J, Pallardo
FV, Mann GE, Vina J (2006) Genistein, a soy isoflavone,
up-regulates expression of antioxidant genes: involvement
of estrogen receptors, erk1/2, and nfkappab. Faseb J
20(12):2136–2138
Boyd AS, Morris LF, Phillips CM, Menter MA (1996) Psori-
asis and pregnancy: hormone and immune system inter-
action. Int J Dermatol 35(3):169–172
Brand RM, Jendrzejewski JL (2008) Topical treatment with
(-)-epigallocatechin-3-gallate and genistein after a single
uv exposure can reduce skin damage. J Dermatol Sci
50(1):69–72
Brandenberger AW, Tee MK, Lee JY, Chao V, Jaffe RB (1997)
Tissue distribution of estrogen receptors alpha (er-alpha)
and beta (er-beta) mrna in the midgestational human fetus.
J Clin Endocrinol Metab 82(10):3509–3512
Braverman IM, Fonferko E (1982) Studies in cutaneous aging:
I. The elastic fiber network. J Invest Dermatol 78(5):
434–443
Brincat MP (2000) Hormone replacement therapy and the skin.
Maturitas 35(2):107–117
Brincat M, Moniz CJ, Studd JW, Darby A, Magos A, Emburey
G, Versi E (1985) Long-term effects of the menopause
and sex hormones on skin thickness. Br J Obstet Gynaecol
92(3):256–259
Brincat M, Versi E, Moniz CF, Magos A, de Trafford J, Studd
JW (1987) Skin collagen changes in postmenopausal
women receiving different regimens of estrogen therapy.
Obstet Gynecol 70(1):123–127
Broue F, Liere P, Kenyon C, Baulieu EE (2007) A steroid
hormone that extends the lifespan of caenorhabditis ele-
gans. Aging Cell 6(1):87–94
Bullough HF (1947) Epidermal thickness following oestrone
injections in the mouse. Nature 159(4029):101
Bulun SE, Zeitoun K, Sasano H, Simpson ER (1999) Aroma-
tase in aging women. Semin Reprod Endocrinol 17(4):
349–358
Cai Q, Shu XO, Jin F, Dai Q, Wen W, Cheng JR, Gao YT,
Zheng W (2003) Genetic polymorphisms in the estrogen
receptor alpha gene and risk of breast cancer: results from
the shanghai breast cancer study. Cancer Epidemiol Bio-
markers Prev 12(9):853–859
Calleja M, Pena P, Ugalde C, Ferreiro C, Marco R, Garesse R
(1993) Mitochondrial DNA remains intact during dro-
sophila aging, but the levels of mitochondrial transcripts are
significantly reduced. J Biol Chem 268(25):18891–18897
Calmels P, Vico L, Alexandre C, Minaire P (1995) Cross-
sectional study of muscle strength and bone mineral
density in a population of 106 women between the ages of
44 and 87 years: relationship with age and menopause.
Eur J Appl Physiol Occup Physiol 70(2):180–186
Cameron IL (1972) Cell proliferation and renewal in aging
mice. J Gerontol 27(2):162–172
Campbell L, Emmerson E, Davies F, Gilliver SC, Krust A,
Chambon P, Ashcroft GS, Hardman MJ (2010) Estrogen
promotes cutaneous wound healing via estrogen receptor
beta independent of it’s anti-inflammatory activities.
J Exp Med 207(9):1825–1833
Carcenac G, Herard ME, Kergoat MJ, Lajeunesse Y, Champ-
oux N, Barsauskas A, Kergoat H (2009) Assessment of
visual function in institutionalized elderly patients. J Am
Med Dir Assoc 10(1):45–49
Cargill SL, Carey JR, Muller HG, Anderson G (2003) Age of
ovary determines remaining life expectancy in old ovari-
ectomized mice. Aging Cell 2(3):185–190
Castelo-Branco C, Duran M, Gonzalez-Merlo J (1992) Skin
collagen changes related to age and hormone replacement
therapy. Maturitas 15(2):113–119
Chakravarti S, Collins WP, Forecast JD, Newton JR, Oram
DH, Studd JW (1976) Hormonal profiles after the meno-
pause. Br Med J 2(6039):784–787
Chen W, Zouboulis CC, Fritsch M, Blume-Peytavi U, Kodelja
V, Goerdt S, Luu-The V, Orfanos CE (1998) Evidence of
heterogeneity and quantitative differences of the type 1
5alpha-reductase expression in cultured human skin
cells—evidence of its presence in melanocytes. J Invest
Dermatol 110(1):84–89
Chen W, Thiboutot D, Zouboulis CC (2002) Cutaneous
androgen metabolism: basic research and clinical per-
spectives. J Invest Dermatol 119(5):992–1007
Chiu TM, Huang CC, Lin TJ, Fang JY, Wu NL, Hung CF
(2009) In vitro and in vivo anti-photoaging effects of an
Biogerontology
123
isoflavone extract from soybean cake. J Ethnopharmacol
126(1):108–113
Cho JL, Allanson M, Domanski D, Arun SJ, Reeve VE (2008)
Estrogen receptor-beta signaling protects epidermal
cytokine expression and immune function from uvb-
induced impairment in mice. Photochem Photobiol Sci
7(1):120–125
Colditz GA, Willett WC, Stampfer MJ, Rosner B, Speizer FE,
Hennekens CH (1987) Menopause and the risk of coro-
nary heart disease in women. N Engl J Med
316(18):1105–1110
Couse JF, Lindzey J, Grandien K, Gustafsson JA, Korach KS
(1997) Tissue distribution and quantitative analysis of
estrogen receptor-alpha (eralpha) and estrogen receptor-
beta (erbeta) messenger ribonucleic acid in the wild-type
and eralpha-knockout mouse. Endocrinology 138(11):
4613–4621
Creidi P, Faivre B, Agache P, Richard E, Haudiquet V, Sau-
vanet JP (1994) Effect of a conjugated oestrogen (prem-
arin) cream on ageing facial skin. A comparative study
with a placebo cream. Maturitas 19(3):211–223
D’Anna R, Cannata ML, Atteritano M, Cancellieri F, Corrado F,
Baviera G, Triolo O, Antico F, Gaudio A, Frisina N, Bitto A,
Polito F, Minutoli L, Altavilla D, Marini H, Squadrito F
(2007) Effects of the phytoestrogen genistein on hot flushes,
endometrium, and vaginal epithelium in postmenopausal
women: a 1-year randomized, double-blind, placebo-con-
trolled study. Menopause 14(4):648–655
De Paula ML, Rodrigues DH, Teixeira HC, Barsante MM,
Souza MA, Ferreira AP (2008) Genistein down-modulates
pro-inflammatory cytokines and reverses clinical signs of
experimental autoimmune encephalomyelitis. Int Immu-
nopharmacol 8(9):1291–1297
Dimri GP, Hara E, Campisi J (1994) Regulation of two e2f-
related genes in presenescent and senescent human
fibroblasts. J Biol Chem 269(23):16180–16186
Dimri GP, Lee X, Basile G, Acosta M, Scott G, Roskelley C,
Medrano EE, Linskens M, Rubelj I, Pereira-Smith O et al
(1995) A biomarker that identifies senescent human cells
in culture and in aging skin in vivo. Proc Natl Acad Sci
USA 92(20):9363–9367
Dumont M, Luu-The V, Dupont E, Pelletier G, Labrie F (1992)
Characterization, expression, and immunohistochemical
localization of 3 beta-hydroxysteroid dehydrogenase/delta
5-delta 4 isomerase in human skin. J Invest Dermatol
99(4):415–421
Dunna SF, Finlay AY (1989) Psoriasis: improvement during
and worsening after pregnancy. Br J Dermatol 120(4):584
Eicheler W, Dreher M, Hoffmann R, Happle R, Aumuller G
(1995) Immunohistochemical evidence for differential
distribution of 5 alpha-reductase isoenzymes in human
skin. Br J Dermatol 133(3):371–376
Emmerson E, Campbell L, Ashcroft GS, Hardman MJ (2009)
Unique and synergistic roles for 17beta-estradiol and
macrophage migration inhibitory factor during cutaneous
wound closure are cell type specific. Endocrinology
150(6):2749–2757
Emmerson E, Campbell L, Ashcroft GS, Hardman MJ (2010)
The phytoestrogen genistein promotes wound healing by
multiple independent mechanisms. Mol Cell Endocrinol
321(2):184–193
Evans WJ (2010) Skeletal muscle loss: cachexia, sarcopenia,
and inactivity. Am J Clin Nutr 91(4):1123S–1127S
Evans JR, Schwartz SD, McHugh JD, Thamby-Rajah Y,
Hodgson SA, Wormald RP, Gregor ZJ (1998) Systemic
risk factors for idiopathic macular holes: a case-control
study. Eye (Lond) 12(Pt 2):256–259
Fiorelli G, Picariello L, Martineti V, Tonelli F, Brandi ML
(1999) Functional estrogen receptor beta in colon cancer
cells. Biochem Biophys Res Commun 261(2):521–527
Flatt T, Min KJ, D’Alterio C, Villa-Cuesta E, Cumbers J,
Lehmann R, Jones DL, Tatar M (2008) Drosophila germ-
line modulation of insulin signaling and lifespan. Proc
Natl Acad Sci USA 105(17):6368–6373
Foley EF, Jazaeri AA, Shupnik MA, Jazaeri O, Rice LW
(2000) Selective loss of estrogen receptor beta in malig-
nant human colon. Cancer Res 60(2):245–248
Frasor J, Stossi F, Danes JM, Komm B, Lyttle CR, Katzenel-
lenbogen BS (2004) Selective estrogen receptor modula-
tors: discrimination of agonistic versus antagonistic
activities by gene expression profiling in breast cancer
cells. Cancer Res 64(4):1522–1533
Fuchs KO, Solis O, Tapawan R, Paranjpe J (2003) The effects
of an estrogen and glycolic acid cream on the facial skin
of postmenopausal women: a randomized histologic
study. Cutis 71(6):481–488
Fukaya S, Iwamoto T, Kin K, Takasaki M (2000) Factors
influencing platelet function in elderly patients with
chronic phase thrombotic diseases. Nippon Ronen Igakkai
Zasshi 37(8):619–626
Gal P, Novotny M, Vasilenko T, Depta F, Sulla I, Tomori Z
(2010) Decrease in wound tensile strength following post-
surgical estrogen replacement therapy in ovariectomized
rats during the early phase of healing is mediated via er-
alpha rather than er-beta: a preliminary report. J Surg Res
159(1):e25–e28
Gates BJ, Sonnett TE, Duvall CA, Dobbins EK (2009) Review
of osteoporosis pharmacotherapy for geriatric patients.
Am J Geriatr Pharmacother 7(6):293–323
Gerisch B, Antebi A (2004) Hormonal signals produced by daf-
9/cytochrome p450 regulate c. Elegans dauer diapause in
response to environmental cues. Development 131(8):
1765–1776
Gerisch B, Weitzel C, Kober-Eisermann C, Rottiers V, Antebi
A (2001) A hormonal signaling pathway influencing c.
Elegans metabolism, reproductive development, and life
span. Dev Cell 1(6):841–851
Giangreco A, Qin M, Pintar JE, Watt FM (2008) Epidermal
stem cells are retained in vivo throughout skin aging.
Aging Cell 7(2):250–259
Gilchrest BA (1983) In vitro assessment of keratinocyte aging.
J Invest Dermatol 81(1 Suppl):184s–189s
Gilchrest BA, Murphy GF, Soter NA (1982a) Effect of
chronologic aging and ultraviolet irradiation on langer-
hans cells in human epidermis. J Invest Dermatol
79(2):85–88
Gilchrest BA, Stoff JS, Soter NA (1982b) Chronologic aging
alters the response to ultraviolet-induced inflammation in
human skin. J Invest Dermatol 79(1):11–15
Gilchrest BA, Szabo G, Flynn E, Goldwyn RM (1983) Chro-
nologic and actinically induced aging in human facial
skin. J Invest Dermatol 80 Suppl:81s–85s
Biogerontology
123
Gilliver SC, Wu F, Ashcroft GS (2003) Regulatory roles of
androgens in cutaneous wound healing. Thromb Haemost
90(6):978–985
Gilliver SC, Ashworth JJ, Mills SJ, Hardman MJ, Ashcroft GS
(2006) Androgens modulate the inflammatory response
during acute wound healing. J Cell Sci 119(Pt 4):722–732
Gilliver SC, Ruckshanthi JP, Hardman MJ, Zeef LA, Ashcroft
GS (2009) 5alpha-dihydrotestosterone (dht) retards wound
closure by inhibiting re-epithelialization. J Pathol
217(1):73–82
Goldzieher JW (1949) The direct effect of steroids on the
senile human skin. J Gerontol 4(2):104–112
Gragnani A, Warde M, Furtado F, Ferreira LM (2009) Topical
tamoxifen therapy in hypertrophic scars or keloids in
burns. Arch Dermatol Res 302(1):1–4
Gurwitz JH (2005) The age/gender interface in geriatric
pharmacotherapy. J Womens Health (Larchmt) 14(1):
68–72
Haratake A, Uchida Y, Mimura K, Elias PM, Holleran WM
(1997) Intrinsically aged epidermis displays diminished
uvb-induced alterations in barrier function associated with
decreased proliferation. J Invest Dermatol 108(3):
319–323
Hardman MJ, Ashcroft GS (2008) Estrogen, not intrinsic aging,
is the major regulator of delayed human wound healing in
the elderly. Genome Biol 9(5):R80
Hardman MJ, Waite A, Zeef L, Burow M, Nakayama T,
Ashcroft GS (2005) Macrophage migration inhibitory
factor: a central regulator of wound healing. Am J Pathol
167(6):1561–1574
Hardman MJ, Emmerson E, Campbell L, Ashcroft GS (2008)
Selective estrogen receptor modulators accelerate cuta-
neous wound healing in ovariectomized female mice.
Endocrinology 149(2):551–557
Harley CB (1991) Telomere loss: mitotic clock or genetic time
bomb? Mutat Res 256(2–6):271–282
Harman D (1956) Aging: a theory based on free radical and
radiation chemistry. J Gerontol 11(3):298–300
Havlikova H, Hill M, Hampl R, Starka L (2002) Sex- and age-
related changes in epitestosterone in relation to pregnen-
olone sulfate and testosterone in normal subjects. J Clin
Endocrinol Metab 87(5):2225–2231
Hazelton GA, Lang CA (1984) Glutathione levels during the
mosquito life span with emphasis on senescence. Proc Soc
Exp Biol Med 176(3):249–256
Hederstierna C, Hultcrantz M, Collins A, Rosenhall U (2007)
Hearing in women at menopause. Prevalence of hearing
loss, audiometric configuration and relation to hormone
replacement therapy. Acta Otolaryngol 127(2):149–155
Hederstierna C, Hultcrantz M, Collins A, Rosenhall U (2010)
The menopause triggers hearing decline in healthy
women. Hear Res 259(1–2):31–35
Henneman DH (1968) Effect of estrogen on in vivo and in vitro
collagen biosynthesis and maturation in old and young
female guinea pigs. Endocrinology 83(4):678–690
Herndon JG, Tigges J, Anderson DC, Klumpp SA, McClure
HM (1999) Brain weight throughout the life span of the
chimpanzee. J Comp Neurol 409(4):567–572
Herrick S, Ashcroft G, Ireland G, Horan M, McCollum C,
Ferguson M (1997) Up-regulation of elastase in acute
wounds of healthy aged humans and chronic venous leg
ulcers are associated with matrix degradation. Lab Invest
77(3):281–288
Herynk MH, Fuqua SA (2004) Estrogen receptor mutations in
human disease. Endocr Rev 25(6):869–898
Hill MW (1988) Influence of age on the morphology and
transit time of murine stratified squamous epithelia. Arch
Oral Biol 33(4):221–229
Holzenberger M, Dupont J, Ducos B, Leneuve P, Geloen A,
Even PC, Cervera P, Le Bouc Y (2003) Igf-1 receptor
regulates lifespan and resistance to oxidative stress in
mice. Nature 421(6919):182–187
Hosono R, Nishimoto S, Kuno S (1989) Alterations of life span
in the nematode caenorhabditis elegans under monoxenic
culture conditions. Exp Gerontol 24(3):251–264
Hsin H, Kenyon C (1999) Signals from the reproductive system
regulate the lifespan of c. Elegans. Nature 399(6734):
362–366
Hu SK, Mitcho YL, Rath NC (1988) Effect of estradiol on
interleukin 1 synthesis by macrophages. Int J Immuno-
pharmacol 10(3):247–252
Hughes SV, Robinson E, Bland R, Lewis HM, Stewart PM,
Hewison M (1997) 1,25-dihydroxyvitamin d3 regulates
estrogen metabolism in cultured keratinocytes. Endocri-
nology 138(9):3711–3718
Humphries S, Stevens DJ (2001) Reproductive biology. Out
with a bang. Nature 410(6830):758–759
Imanishi T, Hano T, Nishio I (2005a) Estrogen reduces
angiotensin ii-induced acceleration of senescence in
endothelial progenitor cells. Hypertens Res 28(3):263–
271
Imanishi T, Hano T, Nishio I (2005b) Estrogen reduces endo-
thelial progenitor cell senescence through augmentation of
telomerase activity. J Hypertens 23(9):1699–1706
Iversen OH, Schjoelberg AR (1984) Age-related changes of
epidermal cell kinetics in the hairless mouse. Virchows
Arch B Cell Pathol Incl Mol Pathol 46(1–2):135–143
Jacobsen BK, Nilssen S, Heuch I, Kvale G (1997) Does age at
natural menopause affect mortality from ischemic heart
disease? J Clin Epidemiol 50(4):475–479
Jia K, Albert PS, Riddle DL (2002) Daf-9, a cytochrome p450
regulating c. Elegans larval development and adult lon-
gevity. Development 129(1):221–231
Kenyon C, Chang J, Gensch E, Rudner A, Tabtiang R (1993) A
c. Elegans mutant that lives twice as long as wild type.
Nature 366(6454):461–464
Kimura KD, Tissenbaum HA, Liu Y, Ruvkun G (1997) Daf-2,
an insulin receptor-like gene that regulates longevity and
diapause in caenorhabditis elegans. Science 277(5328):
942–946
Klass MR (1977) Aging in the nematode caenorhabditis ele-
gans: major biological and environmental factors influ-
encing life span. Mech Ageing Dev 6(6):413–429
Kligman AM, Koblenzer C (1997) Demographics and psy-
chological implications for the aging population. Derma-
tol Clin 15(4):549–553
Kovacs EJ, Faunce DE, Ramer-Quinn DS, Mott FJ, Dy PW,
Frazier-Jessen MR (1996) Estrogen regulation of je/mcp-1
mrna expression in fibroblasts. J Leukoc Biol 59(4):
562–568
Biogerontology
123
Krunic D, Moshir S, Greulich-Bode KM, Figueroa R, Cerezo A,
Stammer H, Stark HJ, Gray SG, Nielsen KV, Hartschuh W,
Boukamp P (2009) Tissue context-activated telomerase in
human epidermis correlates with little age-dependent telo-
mere loss. Biochim Biophys Acta 1792(4):297–308
Ku HH, Brunk UT, Sohal RS (1993) Relationship between
mitochondrial superoxide and hydrogen peroxide pro-
duction and longevity of mammalian species. Free Radic
Biol Med 15(6):621–627
Kuiper GG, Enmark E, Pelto-Huikko M, Nilsson S, Gustafsson
JA (1996) Cloning of a novel receptor expressed in rat
prostate and ovary. Proc Natl Acad Sci USA 93(12):
5925–5930
Kuiper GG, Carlsson B, Grandien K, Enmark E, Haggblad J,
Nilsson S, Gustafsson JA (1997) Comparison of the ligand
binding specificity and transcript tissue distribution of
estrogen receptors alpha and beta. Endocrinology 138(3):
863–870
Kukull WA, Higdon R, Bowen JD, McCormick WC, Teri L,
Schellenberg GD, van Belle G, Jolley L, Larson EB
(2002) Dementia and alzheimer disease incidence: a
prospective cohort study. Arch Neurol 59(11):1737–1746
Labrie F, Belanger A, Cusan L, Gomez JL, Candas B (1997)
Marked decline in serum concentrations of adrenal c19
sex steroid precursors and conjugated androgen metabo-
lites during aging. J Clin Endocrinol Metab 82(8):
2396–2402
Labrie F, Martel C, Balser J (2011) Wide distribution of the
serum dehydroepiandrosterone and sex steroid levels in
postmenopausal women: role of the ovary? Menopause
18(1):30–43
Lane MA, Ball SS, Ingram DK, Cutler RG, Engel J, Read V,
Roth GS (1995) Diet restriction in rhesus monkeys lowers
fasting and glucose-stimulated glucoregulatory end points.
Am J Physiol 268(5 Pt 1):E941–E948
Larsen PL, Albert PS, Riddle DL (1995) Genes that regulate
both development and longevity in caenorhabditis ele-
gans. Genetics 139(4):1567–1583
Lavker RM (1979) Structural alterations in exposed and
unexposed aged skin. J Invest Dermatol 73(1):59–66
Lenhardt R, Hopf HW, Marker E, Akca O, Kurz A, Sche-
uenstuhl H, Sessler DI (2000) Perioperative collagen
deposition in elderly and young men and women. Arch
Surg 135(1):71–74
Liang H, Masoro EJ, Nelson JF, Strong R, McMahan CA,
Richardson A (2003) Genetic mouse models of extended
lifespan. Exp Gerontol 38(11–12):1353–1364
Lin SJ, Kaeberlein M, Andalis AA, Sturtz LA, Defossez PA,
Culotta VC, Fink GR, Guarente L (2002) Calorie
restriction extends saccharomyces cerevisiae lifespan by
increasing respiration. Nature 418(6895):344–348
Lindblad U, Langer RD, Wingard DL, Thomas RG, Barrett-
Connor EL (2001) Metabolic syndrome and ischemic
heart disease in elderly men and women. Am J Epidemiol
153(5):481–489
Lindstedt E, Sandblom P (1975) Wound healing in man: tensile
strength of healing wounds in some patient groups. Ann
Surg 181(6):842–846
Ling S, Zhou L, Li H, Dai A, Liu JP, Komesaroff PA, Sudhir K
(2006) Effects of 17beta-estradiol on growth and apop-
tosis in human vascular endothelial cells: influence of
mechanical strain and tumor necrosis factor-alpha. Ste-
roids 71(9):799–808
Loeser AA (1937) The resorption and action of follicular
hormone rubbed into the skin. J Obstet Gynaecol Br Emp
44:710
Lokkegaard E, Jovanovic Z, Heitmann BL, Keiding N, Ottesen
B, Pedersen AT (2006) The association between early
menopause and risk of ischaemic heart disease: influence
of hormone therapy. Maturitas 53(2):226–233
Longcope C (1971) Metabolic clearance and blood production
rates of estrogens in postmenopausal women. Am J Obstet
Gynecol 111(6):778–781
Lopez-Torres M, Perez-Campo R, Rojas C, Cadenas S, Barja G
(1993) Maximum life span in vertebrates: relationship
with liver antioxidant enzymes, glutathione system,
ascorbate, urate, sensitivity to peroxidation, true mal-
ondialdehyde, in vivo H
2
O
2
, and basal and maximum
aerobic capacity. Mech Ageing Dev 70(3):177–199
Lundgren D (1973) Influence of estrogen and progesterone on
exudation, inflammatory cell migration and granulation
tissue formation in preformed cavities. Scand J Plast
Reconstr Surg 7(1):10–14
Lye M, Donnellan C (2000) Heart disease in the elderly. Heart
84(5):560–566
Magnusson U, Einarsson S (1990) Effects of exogenous oest-
radiol on the number and functional capacity of circulat-
ing mononuclear and polymorphonuclear leukocytes in
the sow. Vet Immunol Immunopathol 25(3):235–247
Mair W, Goymer P, Pletcher SD, Partridge L (2003) Demog-
raphy of dietary restriction and death in drosophila. Sci-
ence 301(5640):1731–1733
Mak HY, Ruvkun G (2004) Intercellular signaling of repro-
ductive development by the c. Elegans daf-9 cytochrome
p450. Development 131(8):1777–1786
Makrantonaki E, Vogel K, Fimmel S, Oeff M, Seltmann H,
Zouboulis CC (2008) Interplay of igf-i and 17beta-estra-
diol at age-specific levels in human sebocytes and fibro-
blasts in vitro. Exp Gerontol 43(10):939–946
Margolis DJ, Knauss J, Bilker W (2002) Hormone replacement
therapy and prevention of pressure ulcers and venous leg
ulcers. Lancet 359(9307):675–677
Marini H, Polito F, Altavilla D, Irrera N, Minutoli L, Calo M,
Adamo EB, Vaccaro M, Squadrito F, Bitto A (2010)
Genistein aglycone improves skin repair in an incisional
model of wound healing: a comparison with raloxifene
and oestradiol in ovariectomized rats. Br J Pharmacol
160(5):1185–1194
Mason JB, Cargill SL, Anderson GB, Carey JR (2009)
Transplantation of young ovaries to old mice increased
life span in transplant recipients. J Gerontol A Biol Sci
Med Sci 64(12):1207–1211
Matsumoto T (2006) Selective estrogen receptor modulators
(serms). Clin Calcium 16(9):100–105
McCay CM, Crowell MF, Maynard LA (1935) The effect of
retarded growth upon the length of the lifespan and upon
the ultimate body size. J Nutr 10:63–79
McCulloch D, Gems D (2003) Evolution of male longevity
bias in nematodes. Aging Cell 2(3):165–173
Melton LJ III, Chrischilles EA, Cooper C, Lane AW, Riggs BL
(1992) Perspective. How many women have osteoporosis?
J Bone Miner Res 7(9):1005–1010
Biogerontology
123
Mendoza CB Jr, Postlethwait RW, Johnson WD (1970) Vet-
erans administration cooperative study of surgery for
duodenal ulcer. Ii. Incidence of wound disruption fol-
lowing operation. Arch Surg 101(3):396–398
Mets T, Bekaert E, Verdonk G (1983) Similarity between in
vitro and in vivo cellular aging. Mech Ageing Dev
22(1):71–78
Miller JG, Mac Neil S (1997) Gender and cutaneous mela-
noma. Br J Dermatol 136(5):657–665
Mills SJ, Ashworth JJ, Gilliver SC, Hardman MJ, Ashcroft GS
(2005) The sex steroid precursor dhea accelerates cuta-
neous wound healing via the estrogen receptors. J Invest
Dermatol 125(5):1053–1062
Miquel J, Economos AC, Fleming J, Johnson JE Jr (1980)
Mitochondrial role in cell aging. Exp Gerontol
15(6):575–591
Mitchell RE (1967) Chronic solar dermatosis: a light and
electron microscopic study of the dermis. J Invest Der-
matol 48(3):203–220
Monteiro-Riviere NA, Banks YB, Birnbaum LS (1991) Laser
Doppler measurements of cutaneous blood flow in ageing
mice and rats. Toxicol Lett 57(3):329–338
Moraes AB, Haidar MA, Soares JMJ, Simoes MJ, Baracat EC,
Patriarca MT (2009) The effects of topical isoflavones on
postmenopausal skin: double-blind and randomized clin-
ical trial of efficacy. Eur J Obstet Gynecol Reprod Biol
146:188–192
Morales DE, McGowan KA, Grant DS, Maheshwari S, Bhar-
tiya D, Cid MC, Kleinman HK, Schnaper HW (1995)
Estrogen promotes angiogenic activity in human umbili-
cal vein endothelial cells in vitro and in a murine model.
Circulation 91(3):755–763
Morin GB (1989) The human telomere terminal transferase
enzyme is a ribonucleoprotein that synthesizes ttaggg
repeats. Cell 59(3):521–529
Morris GM, van den Aardweg GJ, Hamlet R, Whitehouse E,
Hopewell JW, Franke H, Loeffler M (1990) Age-related
changes in the cell kinetics of rat foot epidermis. Cell
Tissue Kinet 23(2):113–123
Morrison JH, Brinton RD, Schmidt PJ, Gore AC (2006)
Estrogen, menopause, and the aging brain: how basic
neuroscience can inform hormone therapy in women.
J Neurosci 26(41):10332–10348
Mosselman S, Polman J, Dijkema R (1996) Er beta: identifi-
cation and characterization of a novel human estrogen
receptor. FEBS Lett 392(1):49–53
Motola DL, Cummins CL, Rottiers V, Sharma KK, Li T, Li Y,
Suino-Powell K, Xu HE, Auchus RJ, Antebi A, Man-
gelsdorf DJ (2006) Identification of ligands for daf-12 that
govern dauer formation and reproduction in c. Elegans.
Cell 124(6):1209–1223
Mousavi SR, Raaiszadeh M, Aminseresht M, Behjoo S (2010)
Evaluating tamoxifen effect in the prevention of hyper-
trophic scars following surgical incisions. Dermatol Surg
36(5):665–669
Nyman S (1971) Studies on the influence of estradiol and
progesterone on granulation tissue. J Periodontal Res
Suppl 7:1–24
Ogawa S, Inoue S, Watanabe T, Hiroi H, Orimo A, Hosoi T,
Ouchi Y, Muramatsu M (1998) The complete primary
structure of human estrogen receptor beta (her beta) and
its heterodimerization with er alpha in vivo and in vitro.
Biochem Biophys Res Commun 243(1):122–126
Onoe Y, Miyaura C, Ohta H, Nozawa S, Suda T (1997)
Expression of estrogen receptor beta in rat bone. Endo-
crinology 138(10):4509–4512
Owsley C, McGwin G, Scilley K, Meek GC, Dyer A, Seker D
(2007) The visual status of older persons residing in
nursing homes. Arch Ophthalmol 125(7):925–930
Pickar JH, Macneil T, Ohleth K (2010) Serms: progress and
future perspectives. Maturitas 67(2):129–138
Pierard-Franchimont C, Letawe C, Goffin V, Pierard GE
(1995) Skin water-holding capacity and transdermal
estrogen therapy for menopause: a pilot study. Maturitas
22(2):151–154
Pillai S, Oresajo C, Hayward J (2005) Ultraviolet radiation and
skin aging: roles of reactive oxygen species, inflammation
and protease activation, and strategies for prevention of
inflammation-induced matrix degradation—a review. Int J
Cosmet Sci 27(1):17–34
Pinto RE, Bartley W (1968) Changes in glutathione reductase
and glutathione peroxidase activities in rat liver related to
age and sex. Biochem J 109(3):34P
Pinto RE, Bartley W (1969) The effect of age and sex on
glutathione reductase and glutathione peroxidase activities
and on aerobic glutathione oxidation in rat liver homog-
enates. Biochem J 112(1):109–115
Pletcher SD, Macdonald SJ, Marguerie R, Certa U, Stearns SC,
Goldstein DB, Partridge L (2002) Genome-wide transcript
profiles in aging and calorically restricted drosophila
melanogaster. Curr Biol 12(9):712–723
Pochi PE, Strauss JS, Downing DT (1979) Age-related changes in
sebaceous gland activity. J Invest Dermatol 73(1):108–111
Punnonen R (1971) On the effect of castration and peroral
estrogen therapy on the skin. Acta Obstet Gynecol Scand
9(Suppl 9):32
Punnonen R, Vaajalahti P, Teisala K (1987) Local oestriol
treatment improves the structure of elastic fibers in the skin
of postmenopausal women. Ann Chir Gynaecol Suppl
202:39–41
Qiu Y, Waters CE, Lewis AE, Langman MJ, Eggo MC (2002)
Oestrogen-induced apoptosis in colonocytes expressing
oestrogen receptor beta. J Endocrinol 174(3):369–377
Rajabi MA, Rajabi F (2007) The effect of estrogen on wound
healing in rats. Pak J Med Sci 23(3):349–352
Ramsey JJ, Colman RJ, Binkley NC, Christensen JD, Gresl
TA, Kemnitz JW, Weindruch R (2000) Dietary restriction
and aging in rhesus monkeys: the University of Wisconsin
study. Exp Gerontol 35(9–10):1131–1149
Reed GA, Tawfik O, Casparian JM (2005) Immunohisto-
chemical analysis of steroid hormone receptors and
structural proteins in skin: effects of aging and hormone
status. J Appl Res 5(2):371–377
Rezzi S, Martin FP, Shanmuganayagam D, Colman RJ, Nich-
olson JK, Weindruch R (2009) Metabolic shifts due to
long-term caloric restriction revealed in nonhuman pri-
mates. Exp Gerontol 44(5):356–362
Richard S, Querleux B, Bittoun J, Jolivet O, Idy-Peretti I, de
Lacharriere O, Leveque JL (1993) Characterization of the
skin in vivo by high resolution magnetic resonance
imaging: water behavior and age-related effects. J Invest
Dermatol 100(5):705–709
Biogerontology
123
Sadoun E, Reed MJ (2003) Impaired angiogenesis in aging is
associated with alterations in vessel density, matrix
composition, inflammatory response, and growth factor
expression. J Histochem Cytochem 51(9):1119–1130
Sadowska-Krowicka H, Mannick EE, Oliver PD, Sandoval M,
Zhang XJ, Eloby-Childess S, Clark DA, Miller MJ (1998)
Genistein and gut inflammation: role of nitric oxide. Proc
Soc Exp Biol Med 217(3):351–357
Sakuraoka K, Tajima S, Seyama Y, Teramoto K, Ishibashi M
(1996) Analysis of connective tissue macromolecular
components in ishibashi rat skin: role of collagen and
elastin in cutaneous aging. J Dermatol Sci 12(3):232–237
Sandblom P, Petersen P, Muren A (1953) Determination of the
tensile strength of the healing wound as a clinical test.
Acta Chir Scand 105(1–4):252–257
Sanz A, Hiona A, Kujoth GC, Seo AY, Hofer T, Kouwenhoven
E, Kalani R, Prolla TA, Barja G, Leeuwenburgh C (2007)
Evaluation of sex differences on mitochondrial bioener-
getics and apoptosis in mice. Exp Gerontol 42(3):173–182
Sastre J, Pallardo FV, Vina J (2000) Mitochondrial oxidative
stress plays a key role in aging and apoptosis. IUBMB
Life 49(5):427–435
Sato R, Maesawa C, Fujisawa K, Wada K, Oikawa K, Takik-
awa Y, Suzuki K, Oikawa H, Ishikawa K, Masuda T
(2004) Prevention of critical telomere shortening by
oestradiol in human normal hepatic cultured cells and
carbon tetrachloride induced rat liver fibrosis. Gut
53(7):1001–1009
Savvas M, Bishop J, Laurent G, Watson N, Studd J (1993)
Type iii collagen content in the skin of postmenopausal
women receiving oestradiol and testosterone implants. Br
J Obstet Gynaecol 100(2):154–156
Sawaya ME, Penneys NS (1992) Immunohistochemical dis-
tribution of aromatase and 3b-hydroxysteroid dehydro-
genase in human hair follicle and sebaceous gland.
J Cutan Pathol 19(4):309–314
Schmidt JB, Binder M, Macheiner W, Kainz C, Gitsch G,
Bieglmayer C (1994) Treatment of skin ageing symptoms
in perimenopausal females with estrogen compounds. A
pilot study. Maturitas 20(1):25–30
Schmidt JB, Binder M, Demschik G, Bieglmayer C, Reiner A
(1996) Treatment of skin aging with topical estrogens. Int
J Dermatol 35(9):669–674
Schneider EL, Mitsui Y (1976) The relationship between in
vitro cellular aging and in vivo human age. Proc Natl
Acad Sci USA 73(10):3584–3588
Schroeder P, Gremmel T, Berneburg M, Krutmann J (2008)
Partial depletion of mitochondrial DNA from human skin
fibroblasts induces a gene expression profile reminiscent
of photoaged skin. J Invest Dermatol 128(9):2297–2303
Seibel J, Molzberger AF, Hertrampf T, Laudenbach-Les-
chowski U, Diel P (2009) Oral treatment with genistein
reduces the expression of molecular and biochemical
markers of inflammation in a rat model of chronic tnbs-
induced colitis. Eur J Nutr 48(4):213–220
Selman C, Lingard S, Choudhury AI, Batterham RL, Claret M,
Clements M, Ramadani F, Okkenhaug K, Schuster E,
Blanc E, Piper MD, Al-Qassab H, Speakman JR, Carmi-
gnac D, Robinson IC, Thornton JM, Gems D, Partridge L,
Withers DJ (2008) Evidence for lifespan extension and
delayed age-related biomarkers in insulin receptor sub-
strate 1 null mice. FASEB J 22(3):807–818
Shaw TJ, Martin P (2009) Wound repair at a glance. J Cell Sci
122(Pt 18):3209–3213
Shuster S, Black MM, McVitie E (1975) The influence of age
and sex on skin thickness, skin collagen and density. Br J
Dermatol 93(6):639–643
Shyong EQ, Lu Y, Lazinsky A, Saladi RN, Phelps RG, Austin
LM, Lebwohl M, Wei H (2002) Effects of the isoflavone
40,5,7-trihydroxyisoflavone (genistein) on psoralen plus
ultraviolet a radiation (puva)-induced photodamage. Car-
cinogenesis 23(2):317–321
Siesky BA, Harris A, Patel C, Klaas CL, Harris M, McCranor
LJ, Lauer J, Kaplan B (2008) Comparison of visual
function and ocular hemodynamics between pre- and post-
menopausal women. Eur J Ophthalmol 18(2):320–323
Simon AF, Shih C, Mack A, Benzer S (2003) Steroid control of
longevity in drosophila melanogaster. Science 299(5611):
1407–1410
Simonen O, Mikkola T (1991) Senile osteoporosis and femoral
neck fractures in long-stay institutions. Calcif Tissue Int
49 Suppl:S78–S79
Simpkins JW, Yang SH, Wen Y, Singh M (2005)
Estrogens, progestins, menopause and neurodegenera-
tion: basic and clinical studies. Cell Mol Life Sci
62(3):271–280
Simpson ER, Zhao Y, Agarwal VR, Michael MD, Bulun SE,
Hinshelwood MM, Graham-Lorence S, Sun T, Fisher CR,
Qin K, Mendelson CR (1997) Aromatase expression in
health and disease. Recent Prog Horm Res 52:185–213
(discussion 213–184)
Smith QT, Allison DJ (1966) Changes of collagen content in
skin, femur and uterus of 17-beta-estradiol benzoate-
treated rats. Endocrinology 79(3):486–492
Sobel H, Lee KD, Hewlett MJ (1965) Effect of estrogen on
acid glycosaminoglycans in skin of mice. Biochim Bio-
phys Acta 101(2):225–229
Son ED, Lee JY, Lee S, Kim MS, Lee BG, Chang IS, Chung
JH (2005) Topical application of 17beta-estradiol increa-
ses extracellular matrix protein synthesis by stimulating
tgf-beta signaling in aged human skin in vivo. J Invest
Dermatol 124(6):1149–1161
Stanford JL, Hartge P, Brinton LA, Hoover RN, Brookmeyer R
(1987) Factors influencing the age at natural menopause.
J Chronic Dis 40(11):995–1002
Stanulis-Praeger BM, Gilchrest BA (1986) Growth factor
responsiveness declines during adulthood for human skin-
derived cells. Mech Ageing Dev 35(2):185–198
Stanulis-Praeger BM, Gilchrest BA (1989) Effect of donor age
and prior sun exposure on growth inhibition of cultured
human dermal fibroblasts by all trans-retinoic acid. J Cell
Physiol 139(1):116–124
Stevenson S, Sharpe DT, Thornton MJ (2009) Effects of
oestrogen agonists on human dermal fibroblasts in an in
vitro wounding assay. Exp Dermatol 18(11):988–990
Sthoeger ZM, Zinger H, Mozes E (2003) Beneficial effects of
the anti-oestrogen tamoxifen on systemic lupus erythe-
matosus of (nzbxnzw)f1 female mice are associated with
specific reduction of igg3 autoantibodies. Ann Rheum Dis
62(4):341–346
Biogerontology
123
Stindl R (2004) Tying it all together: telomeres, sexual size
dimorphism and the gender gap in life expectancy. Med
Hypotheses 62(1):151–154
Sugimoto M, Yamashita R, Ueda M (2006) Telomere length of
the skin in association with chronological aging and
photoaging. J Dermatol Sci 43(1):43–47
Sumino H, Ichikawa S, Abe M, Endo Y, Ishikawa O, Kura-
bayashi M (2004) Effects of aging, menopause, and hor-
mone replacement therapy on forearm skin elasticity in
women. J Am Geriatr Soc 52(6):945–949
Sumino H, Ichikawa S, Kasama S, Takahashi T, Kumakura H,
Takayama Y, Kanda T, Murakami M, Kurabayashi M
(2009) Effects of raloxifene and hormone replacement
therapy on forearm skin elasticity in postmenopausal
women. Maturitas 62(1):53–57
Surazynski A, Jarzabek K, Haczynski J, Laudanski P, Palka J,
Wolczynski S (2003) Differential effects of estradiol and
raloxifene on collagen biosynthesis in cultured human
skin fibroblasts. Int J Mol Med 12(5):803–809
Suthers K, Kim JK, Crimmins E (2003) Life expectancy with
cognitive impairment in the older population of the United
States. J Gerontol B Psychol Sci Soc Sci 58(3):S179–S186
Taguchi A, Wartschow LM, White MF (2007) Brain irs2 sig-
naling coordinates life span and nutrient homeostasis.
Science 317(5836):369–372
Tatar M, Kopelman A, Epstein D, Tu MP, Yin CM, Garofalo
RS (2001) A mutant drosophila insulin receptor homolog
that extends life-span and impairs neuroendocrine func-
tion. Science 292(5514):107–110
Thiboutot D, Martin P, Volikos L, Gilliland K (1998) Oxidative
activity of the type 2 isozyme of 17beta-hydroxysteroid
dehydrogenase (17beta-hsd) predominates in human seba-
ceous glands. J Invest Dermatol 111(3):390–395
Thiboutot D, Bayne E, Thorne J, Gilliland K, Flanagan J, Shao
Q, Light J, Helm K (2000) Immunolocalization of 5alpha-
reductase isozymes in acne lesions and normal skin. Arch
Dermatol 136(9):1125–1129
Thiers BH, Maize JC, Spicer SS, Cantor AB (1984) The effect
of aging and chronic sun exposure on human langerhans
cell populations. J Invest Dermatol 82(3):223–226
Thomas JR (2005) Effects of age and diet on rat skin histology.
Laryngoscope 115(3):405–411
Thornton MJ, Taylor AH, Mulligan K, Al-Azzawi F, Lyon CC,
O’Driscoll J, Messenger AG (2003a) 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 8(1):100–103
Thornton MJ, Taylor AH, Mulligan K, Al-Azzawi F, Lyon CC,
O’Driscoll J, Messenger AG (2003b) Oestrogen receptor
beta is the predominant oestrogen receptor in human scalp
skin. Exp Dermatol 12(2):181–190
Tian DS, Liu JL, Xie MJ, Zhan Y, Qu WS, Yu ZY, Tang ZP,
Pan DJ, Wang W (2009) Tamoxifen attenuates inflam-
matory-mediated damage and improves functional out-
come after spinal cord injury in rats. J Neurochem
109(6):1658–1667
Tiganescu A, Walker EA, Hardy RS, Mayes AE, Stewart PM
(2011) Localization, age- and site-dependent expression,
and regulation of 11beta-hydroxysteroid dehydrogenase
type 1 in skin. J Invest Dermatol 131(1):30–36
Toutain CE, Brouchet L, Raymond-Letron I, Vicendo P, Ber-
ges H, Favre J, Fouque MJ, Krust A, Schmitt AM,
Chambon P, Gourdy P, Arnal JF, Lenfant F (2009) Pre-
vention of skin flap necrosis by estradiol involves reper-
fusion of a protected vascular network. Circ Res
104(2):245–254 (212p following 254)
Tresguerres JA, Kireev R, Tresguerres AF, Borras C, Vara E,
Ariznavarreta C (2008) Molecular mechanisms involved
in the hormonal prevention of aging in the rat. J Steroid
Biochem Mol Biol 108(3–5):318–326
Trichopoulos D, MacMahon B, Cole P (1972) Menopause and
breast cancer risk. J Natl Cancer Inst 48(3):605–613
Tsoureli-Nikita E, Watson RE, Griffiths CE (2006) Photoage-
ing: the darker side of the sun. Photochem Photobiol Sci
5(2):160–164
Tsukahara K, Moriwaki S, Ohuchi A, Fujimura T, Takema Y
(2001) Ovariectomy accelerates photoaging of rat skin.
Photochem Photobiol 73(5):525–531
Tsukahara K, Nakagawa H, Moriwaki S, Kakuo S, Ohuchi A,
Takema Y, Imokawa G (2004) Ovariectomy is sufficient
to accelerate spontaneous skin ageing and to stimulate
ultraviolet irradiation-induced photoageing of murine
skin. Br J Dermatol 151(5):984–994
Varani J, Spearman D, Perone P, Fligiel SE, Datta SC, Wang
ZQ, Shao Y, Kang S, Fisher GJ, Voorhees JJ (2001)
Inhibition of type i procollagen synthesis by damaged
collagen in photoaged skin and by collagenase-degraded
collagen in vitro. Am J Pathol 158(3):931–942
Varila E, Rantala I, Oikarinen A, Risteli J, Reunala T, Oksanen
H, Punnonen R (1995) The effect of topical oestradiol on
skin collagen of postmenopausal women. Br J Obstet
Gynaecol 102(12):985–989
Verdier-Sevrain S, Yaar M, Cantatore J, Traish A, Gilchrest
BA (2004) Estradiol induces proliferation of keratinocytes
via a receptor mediated mechanism. FASEB J 18(11):
1252–1254
Vina J, Borras C, Gambini J, Sastre J, Pallardo FV (2005) Why
females live longer than males? Importance of the
upregulation of longevity-associated genes by oestrogenic
compounds. FEBS Lett 579(12):2541–2545
Vukelic S, Stojadinovic O, Pastar I, Rabach M, Krzyzanowska
A, Lebrun E, Davis SC, Resnik S, Brem H, Tomic-Canic M
(2011) Cortisol synthesis in epidermis is induced by IL-1
and tissue injury. J Biol Chem. doi:10.1074/jbc.
M110.188268
Waetjen LE, Ye J, Feng WY, Johnson WO, Greendale GA,
Sampselle CM, Sternfield B, Harlow SD, Gold EB (2009)
Association between menopausal transition stages and
developing urinary incontinence. Obstet Gynecol
114(5):989–998
Walter P, Green S, Greene G, Krust A, Bornert JM, Jeltsch JM,
Staub A, Jensen E, Scrace G, Waterfield M et al (1985)
Cloning of the human estrogen receptor cdna. Proc Natl
Acad Sci USA 82(23):7889–7893
Wang E (1995) Senescent human fibroblasts resist pro-
grammed cell death, and failure to suppress bcl2 is
involved. Cancer Res 55(11):2284–2292
Wang YN, Wu W, Chen HC, Fang H (2010) Genistein protects
against uvb-induced senescence-like characteristics in
human dermal fibroblast by p66shc down-regulation.
J Dermatol Sci 58(1):19–27
Biogerontology
123
Weinstein BE, Ventry IM (1982) Hearing impairment and
social isolation in the elderly. J Speech Hear Res
25(4):593–599
Weinstock MA (1994) Epidemiologic investigation of non-
melanoma skin cancer mortality: the Rhode Island follow-
back study. J Invest Dermatol 102(6):6S–9S
Wich SA, Utami-Atmoko SS, Setia TM, Rijksen HD, Schur-
mann C, van Hooff JA, van Schaik CP (2004) Life history
of wild sumatran orangutans (pongo abelii). J Hum Evol
47(6):385–398
Widyarini S, Spinks N, Husband AJ, Reeve VE (2001) Isofl-
avonoid compounds from red clover (trifolium pratense)
protect from inflammation and immune suppression
induced by UV radiation. Photochem Photobiol 74(3):
465–470
Williams ME, Pannill FC III (1982) Urinary incontinence in
the elderly: physiology, pathophysiology, diagnosis, and
treatment. Ann Intern Med 97(6):895–907
Yamawaki TM, Berman JR, Suchanek-Kavipurapu M,
McCormick M, Maria Gaglia M, Lee SJ, Kenyon C
(2010) The somatic reproductive tissues of c. Elegans
promote longevity through steroid hormone signaling.
PLoS Biol 8(8):e1000468
Yancik R, Ganz PA, Varricchio CG, Conley B (2001) Per-
spectives on comorbidity and cancer in older patients:
approaches to expand the knowledge base. J Clin Oncol
19(4):1147–1151
Yang JZ, O’Flatharta C, Harvey BJ, Thomas W (2008)
Membrane eralpha-dependent activation of pkcalpha in
endometrial cancer cells by estradiol. Steroids 73(11):
1110–1122
Zou K, Ing NH (1998) Oestradiol up-regulates oestrogen
receptor, cyclophilin, and glyceraldehyde phosphate
dehydrogenase mRNA concentrations in endometrium,
but down-regulates them in liver. J Steroid Biochem Mol
Biol 64(5–6):231–237
Zouboulis CC, Seltmann H, Hiroi N, Chen W, Young M, Oeff
M, Scherbaum WA, Orfanos CE, McCann SM, Bornstein
SR (2002) Corticotropin-releasing hormone: an autocrine
hormone that promotes lipogenesis in human sebocytes.
Proc Natl Acad Sci USA 99(10):7148–7153
Biogerontology
123
... However, whether advanced age is an isolated risk factor for developing chronic wounds it is not entirely understood. Limited experimental reports suggest that the healing of uncomplicated skin wounds in healthy-aged humans may not be delayed due to intrinsic age but rather to reduced hormone levels (Emmerson and Hardman, 2012). Another research group has found no difference in wound closure between aged and young mice unless they implemented a microbial burden into the wounds (Bagood et al., 2021), which underlines the importance of a multifactorial approach to study differences in woundhealing progression (Bagood et al., 2021). ...
Chapter
Wound healing is a complex and dynamic response to injury which can be divided into three phases: inflammation, re‐epithelialization and matrix remodelling. Interactions are required between keratinocytes, fibroblasts, endothelial cells and inflammatory cells, growth factors, extracellular matrix and enzymes called proteases. Foetal wounds can heal without scarring. However, wounds in children and adults heal with a remodelling phase resulting in a scar. Non‐healing or chronic wounds occur because of infection or pressure as well as systemic factors such as ischaemia or diabetes mellitus. Abnormal scarring results in hypertrophic or keloid scars. Recent advances in stem cell biology and tissue engineering will provide new wound healing therapies.
Article
Full-text available
Documented male-female differences in the risk of cardiovascular and chronic kidney diseases have been largely attributed to estrogens. The cardiovascular and renal protective effects of estrogens are mediated via the activation of estrogen receptors (ERα and ERβ) and G protein-coupled estrogen receptor, and involve interactions with the renin-angiotensin-aldosterone system. Aromatase, also called estrogen synthase, is a cytochrome P-450 enzyme that plays a pivotal role in the conversion of androgens into estrogens. Estrogens are biosynthesized in gonadal and extra-gonadal sites by the action of aromatase. Evidence suggests that aromatase inhibitors, which are used to treat high estrogen–related pathologies, are associated with the development of cardiovascular events. We review the potential role of aromatization in providing cardio-renal protection and highlight several meta-analysis studies on cardiovascular events associated with aromatase inhibitors. Overall, we present the potential of aromatase enzyme as a fundamental contributor to cardio-renal protection.
Article
Full-text available
Non-healing skin wounds are disproportionally prevalent in older adults. Current treatments do not account for the particularities of aged skin and result in inadequate outcomes. Overall, healing chronic wounds in the elderly remains a major unmet clinical need. Estrogens play a critical role in reproduction but also have important actions in non-reproductive organs. Estrogen biosynthesis and signaling pathways are locally activated during physiological wound healing, processes that are inhibited in elderly estrogen-deprived skin. Estrogen deprivation has been shown to be a critical mediator of impaired wound healing in both postmenopausal women and aged men, and topical estrogen application reverses age-associated delayed wound healing in both elderly men and women. These data indicate that adequate estrogen biosynthesis and properly regulated estrogen signaling pathways are essential for normal wound healing and can be targeted to optimize tissue repair in the elderly. However, due to fundamental questions regarding how to safely restore estrogen signaling locally in skin wounds, there are currently no therapeutic strategies addressing estrogen deficiency in elderly chronic wounds. This review discusses established and recent literature in this area and proposes the hypothesis that estrogen plays a pleiotropic role in skin aging and that targeting estrogen signaling and biosynthesis could promote skin repair in older adults.
Article
Full-text available
Cellular senescence is a response to a wide variety of stressors, including DNA damage, oncogene activation and physiologic aging, and pathologically accelerated senescence contributes to human disease, including diabetes mellitus. Indeed, recent work in this field has demonstrated a role for pancreatic β-cell senescence in the pathogenesis of Type 1 Diabetes, Type 2 Diabetes and monogenic diabetes. Small molecule or genetic targeting of senescent β-cells has shown promise as a novel therapeutic approach for preventing and treating diabetes. Despite these advances, major questions remain around the molecular mechanisms driving senescence in the β-cell, identification of molecular markers that distinguish senescent from non-senescent β-cell subpopulations, and translation of proof-of-concept therapies into novel treatments for diabetes in humans. Here, we summarize the current state of the field of β-cell senescence, highlighting insights from mouse models as well as studies on human islets and β-cells. We identify markers that have been used to detect β-cell senescence to unify future research efforts in this field. We discuss emerging concepts of the natural history of senescence in β-cells, heterogeneity of senescent β-cells subpopulations, role of sex differences in senescent responses, and the consequences of senescence on integrated islet function and microenvironment. As a young and developing field, there remain many open research questions which need to be addressed to move senescence-targeted approaches towards clinical investigation.
Chapter
Wound healing is a complex and dynamic response to injury that can be divided into three phases: inflammation, re‐epithelialization and matrix remodelling. Interactions are required between keratinocytes, fibroblasts, endothelial and inflammatory cells, growth factors, extracellular matrix and enzymes called proteases. Fetal wounds can heal without scarring; however wounds in children and adults heal with a remodelling phase resulting in a scar. Non‐healing or chronic wounds occur because of infection or pressure as well as systemic factors such as ischaemia or diabetes. Abnormal scarring results in hypertrophic or keloid scars. The effect of secreted heat shock proteins, regulation by microRNAs, manipulation of macrophage polarization and wound treatment with mesenchymal stem cells (by direct application or mobilization from bone marrow) are new areas of interest.
Chapter
Ovarian aging characterizes menopausal syndrome, which is a dynamic decline in ovarian function and hormone secretion, causing physical and mental symptoms. The menopausal syndrome presentation depends on race, climate, genetics, nutrition, weight, height, birth, and other factors. The average age at which a woman enters perimenopause is 40 years, lasting an average of 4 years but up to 10 years. The root cause of the menopausal syndrome is a decline in ovarian endocrine function, especially estrogen concentration. Primordial follicle depletion accelerates and periodic follicle recruitment and the estrogen concentration decrease starting around the age of 37.5 years. Further, the hypothalamic-pituitary-ovarian (HPO) axis feedback regulation induces an increase in follicle-stimulating hormone (FSH) and a decrease in anti-Müllerian hormone (AMH) and the antral follicle count (AFC).
Chapter
The initial primordial follicle pool size and the activation and depletion rates determine fertility potential. Generally, primordial follicles have three states: resting, growth, and atresia/degeneration. Over-activation and abnormal atresia of primordial follicles are leading causes of ovarian aging [1]. Therefore, this section focuses on the molecular mechanisms of primordial follicle activation (PFA) and follicular atresia.
Article
Full-text available
Studies in invertebrates have led to the identification of a number of genes that regulate lifespan, some of which encode components of the insulin or insulin-like signalling pathways. Examples include the related tyrosine kinase receptors InR (Drosophila melanogaster) and DAF-2 (Caenorhabditis elegans) that are homologues of the mammalian insulin-like growth factor type 1 receptor (IGF-1R). To investigate whether IGF-1R also controls longevity in mammals, we inactivated the IGF-1R gene in mice (Igf1r). Here, using heterozygous knockout mice because null mutants are not viable, we report that Igf1r(+/-) mice live on average 26% longer than their wild-type littermates (P < 0.02). Female Igf1r(+/-) mice live 33% longer than wild-type females (P < 0.001), whereas the equivalent male mice show an increase in lifespan of 16%, which is not statistically significant. Long-lived Igf1r(+/-) mice do not develop dwarfism, their energy metabolism is normal, and their nutrient uptake, physical activity, fertility and reproduction are unaffected. The Igf1r(+/-) mice display greater resistance to oxidative stress, a known determinant of ageing. These results indicate that the IGF-1 receptor may be a central regulator of mammalian lifespan.
Article
Full-text available
When human fibroblasts from different donors are grown in vitro, only a small fraction of the variation in their finite replicative capacity is explae by the chronological age of the donor. Because we had previously shown that telomeres, the terminal guanine-rich sequences of chromosomes , shorten throughout the lifespan of cultured cells, we wished to determine whether variation in initial telomere length would account for the unexplained variation in replicative capacity. Analysis of cells from 31 donors (aged 0-93 yr) indicated relatively weak correlations between proliferative ability and donor age (m =-0.2 doubling per yr; r =-0.42; P = 0.02) and between telomeric DNA and donor age (m =-15 base pairs per yr; r =-0.43; P = 0.02). However, there was a stiking correlation, valid over the entire age range of the donors, between replicative capacity and initial telomere length (m = 10 doublngs per kilobase pair; r = 0.76; P = 0.004), indicating that cell strains with shorter telomeres underwent slglcantiy fewer doublings than those with longer telomeres. These observations suggest that telomere length is a biomarker ofsomatic cell aging in humans and are consistent with a causal role for telomere loss in this process. We also found that fibroblasts from Hutchinson-Gilford progeria donors had short telomeres, consistent with their reduced division potential in vitro. In contrast, telomeres from sperm DNA did not decrease with age ofthe donor, suggesting that a mechanism for maintaining telomere length, such as telomerase expression, may be active in germ-line tissue.
Article
The daf-9 gene functions to integrate transforming growth factor-β and insulin-like signaling pathways to regulate Caenorhabditis elegans larval development. Mutations in daf-9 result in transient dauer-like larval arrest, abnormal reproductive development, molting defects and increased adult longevity. The phenotype is sterol-dependent, and dependent on the activity of DAF-12, a nuclear hormone receptor. Genetic tests show that daf-9 is upstream of daf-12 in the genetic pathways for larval development and adult longevity. daf-9 encodes a cytochrome P450 related to those involved in biosynthesis of steroid hormones in mammals. We propose that it specifies a step in the biosynthetic pathway for a DAF-12 ligand, which might be a steroid. The surprising cellular specificity of daf-9 expression (predominantly in two sensory neurons) supports a previously unrecognized role for these cells in neuroendocrine control of larval development, reproduction and life span.
Article
With the use of the constant infusion technique, the mean metabolic clearance rate for estradiol in 5 women aged 70 to 88 was found to be 580 ± 30 L. per day per square meter. The mean metabolic clearance rate for estrone in 5 women aged 73 to 89 was 1,050 ± 70 L. per day per square meter. Both these represent a decrease of about 25 per cent compared to the corresponding clearance rates of these estrogens in young women. In 14 women aged 73 to 89, the mean plasma concentration of estradiol was 6.5 ± 0.7 pg. per milliliter, and that of estrone was 24.8 ± 3.5 pg. per milliliter. The calculated blood production rate for estradiol in these groups of postmenopausal women was 6 μg per day, and that for estrone was 40 μg per day. Direct secretion of estrogens would appear to be minimal in these asymptomatic postmenopausal women.
Article
Estrogen receptor (ER) is a ligand-activated transcription factor that mediates estrogen actions in target tissues. Several common polymorphisms of the ER-alpha gene have been reported to be associated with alterations in receptor expression and function. We evaluated the hypothesis that genetic polymorphisms in the ER-alpha gene may be associated with breast cancer risk in a population-based case-control study conducted in urban Shanghai during 1996-1998. Two RFLPs at the ER-alpha gene locus, denoted as PvuII and XbaI, were examined in 1069 breast cancer cases and 1166 age frequency-matched controls. PvuII polymorphism was associated with an increased risk of breast cancer with the age-adjusted odds ratios for genotypes Pp and pp being 1.3 [95% confidence interval (CI), 1.0-1.7) and 1.4 (95% CI, 1.1-1.8), respectively, comparing to genotype PP. The XbaI polymorphism was associated with a nonsignificantly elevated risk. The odds ratios for genotypes Xx and xx were 1.2 (95% CI, 0.7-1.9) and 1.3 (95% CI, 0.8-2.0), respectively, and the elevated risks were mainly confined to older or postmenopausal women. No apparent synergetic effect of these two polymorphisms was identified. Results of this study indicate that genetic polymorphisms in the ER-alpha gene may play a role in the etiology of breast cancer.
Article
Epidemiological studies of postmenopausal hormone replacement therapy show a reduction in the risk of developing colon cancer, and animal studies using 17-oestradiol (E 2 ) demonstrate a decreased incidence of chemically-induced colon cancer. Using the colon cancer cell line, COLO205, we found that E 2 induced a dose-dependent increase in DNA fragmentation and nuclear condensation, significant effects being seen at 10 12 mol/l. BSA-conjugated E 2 , which cannot enter cells, was ineffective at inducing apoptosis in COLO205 cells, indicating that E2 was not acting through a cell-membrane receptor. E2 did not induce the morphological changes characteristic of differentiation. Using RT-PCR we found that the oestrogen receptor (ER) isoform was absent in the COLO205 cell line in contrast to CACO-2, LoVo and SW620 cells, but mRNAs for ER1, -2, -5 and -6 isoforms were detected. Western immunoblotting results showed fulllength ER protein but no detectable ER in COLO205 cells. In normal human colon tissue samples immunoreactive ER was found but ER was barely detectable. Expression of ER was lost in some colon cancer specimens and reduced in others. We conclude that E 2 , through ER, at concentrations found during replacement therapy, may inhibit the development of colon cancer by inducing apoptosis.
Article
The healing of test wounds was studied in 108 paitents, in whom some impairment of wound healing was suspected. A 5 cm skin wound was performed in the forearm and the strength of the wound was tested after 5 days using the technique described by Sandblom and associates with two measurements in each wound. No differences in wound strength could be registered between the two wounds in each patient, between males and females nor in patients with malignant disease compared to other patients. Patients with low serum protein or serum albumin values had significantly weaker wounds than patients with normal protein values. Patients over 80 years of age had wounds somewhat weaker than those below 70, the difference having a statistical significance of 6%. The wound strenght in patients was compared to values found elsewhere for wounds in rabbits, rats, and piglets. The pigs had much higher values than others, rabbits slightly stronger than and rats about equal to humans.