Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Sports Med

DOI 10.1007/s40279-013-0081-6

REVIEW ARTICLE

How Sex Hormones Promote Skeletal Muscle Regeneration


Martina Velders • Patrick Diel

! Springer International Publishing Switzerland 2013

Abstract Skeletal muscle regeneration efficiency controlled human studies is presented to support the notion
declines with age for both men and women. This decline that hormonal therapies and exercise induce added positive
impacts on functional capabilities in the elderly and limits effects on functional measures and lean tissue mass. Based
their ability to engage in regular physical activity and to on the fact that aging human skeletal muscle retains the
maintain independence. Aging is associated with a decline ability to adapt to exercise with enhanced satellite cell
in sex hormone production. Therefore, elucidating the activation, combining sex hormone therapies with exercise
effects of sex hormone substitution on skeletal muscle may induce additive effects on satellite cell accretion.
homeostasis and regeneration after injury or disuse is There is evidence to suggest that there is a ‘window of
highly relevant for the aging population, where sarcopenia opportunity’ after the onset of a hypogonadal state such as
affects more than 30 % of individuals over 60 years of age. menopause, to initiate a hormonal therapy in order to
While the anabolic effects of androgens are well known, achieve maximal benefits for skeletal muscle health. Novel
the effects of estrogens on skeletal muscle anabolism have receptor subtype selective ligands and selective estrogen
only been uncovered in recent times. Hence, the purpose of and androgen receptor modulators (SERMs, SARMs)
this review is to provide a mechanistic insight into the promise to reduce health risks associated with classical
regulation of skeletal muscle regenerative processes by hormonal therapies, whilst maintaining the positive effects
both androgens and estrogens. Animal studies using on muscle repair. Dietary supplements containing com-
estrogen receptor (ER) antagonists and receptor subtype pounds with structural similarity to estrogens (phytoestro-
selective agonists have revealed that estrogens act through gens) are increasingly used as alternatives to classical
both genomic and non-genomic pathways to reduce leu- hormone-replacement therapies (HRT), but the effects on
kocyte invasion and increase satellite cell numbers in skeletal muscle are currently largely unknown. Research
regenerating skeletal muscle tissue. Although animal has started to investigate the combined effects of exercise
studies have been more conclusive than human studies in and alternative HRTs, such as soy isoflavones, on skeletal
establishing a role for sex hormones in the attenuation of muscle regenerative processes to provide safer and more
muscle damage, data from a number of recent well efficient therapies to promote muscle regeneration and
maintenance of muscle mass and strength in the aging
population.
M. Velders (&)
Division of Sports Medicine, Department of Internal Medicine,
University Hospital Ulm, Leimgrubenweg 12-14,
1 Introduction
89075 Ulm, Germany
e-mail: martina.velders@gmail.com; martina.velders@uniklinik-
ulm.de Efficient skeletal muscle regeneration is an important
aspect for the aging population, where declining muscle
P. Diel
mass and strength impacts on the ability of older people to
Department of Molecular and Cellular Sports Medicine,
Institute of Sports Medicine, German Sports University, engage in regular physical activity and increases the inci-
Cologne, Germany dence of frailty-related falls. The blunted satellite cell
M. Velders, P. Diel

response in skeletal muscle of old versus young men fol- regeneration in rodent injury, disuse or exercise models.
lowing eccentric exercise is evidence for a regeneration There is considerably less evidence available on the effects
deficit of aging muscle [1]. Early skeletal muscle cross-age of hormonal status on human skeletal muscle regeneration,
transplantation experiments have revealed that muscle but existing literature is reviewed and compared with
regeneration is superior when an old muscle is transplanted findings from animal studies. In the last part of this review,
into a young organism compared with vice versa [2]. This we examine data on the effects of estrogens and androgens
research first implicated a role for circulating factors as therapeutic agents and discuss how this relates to exer-
associated with the host environment in regenerative pro- cise training in the elderly population.
cesses. A large body of literature, mainly from rodent
studies, now provides strong evidence for the ability of 1.1 Skeletal Muscle Regeneration: An Overview
androgenic and estrogenic sex hormones to augment
satellite cell activation and to modulate inflammatory Although skeletal muscle is a post-mitotic tissue, it pos-
dynamics during muscle regeneration [3–7]. Sarcopenia is sesses a remarkable capacity for regeneration after injury
a major health issue, affecting 30 % of individuals over or disuse. This is evidenced by results from a rodent study
60 years of age, leading to falls and physical disability. showing that a whole muscle can be removed, minced and
However, aging skeletal muscle of both men and women replaced, with morphological characteristics being restored
has been shown to retain the ability to respond to exercise within 30–40 days [9]. The skeletal muscle repair process
with increased satellite cell numbers [8]. Since estrogens can be categorized into three parts: (i) necrosis, (ii)
have been shown to increase satellite cell numbers [5], phagocytosis and repair, and (iii) remodelling and resto-
combining exercise with pharmacological hormone thera- ration of muscle function [10]. The initial event after
pies seems a promising treatment strategy to increase muscle injury is necrosis due to membrane disruption and
muscle mass and improve regeneration in elderly people. activation of calcium-dependent proteases [11]. This is
Elucidating the molecular targets activated by the combi- followed by an invasion of myeloid immune cells into the
natory influence of both exercise and sex hormones in injured skeletal muscle areas. Myeloid cells numbers can
skeletal muscle is essential in order to design effective exceed 100,000 cells/mm3 in injured postnatal muscle [12].
strategies to counteract sarcopenia and improve muscle Neutrophils are the first immune cells to be recruited to the
regeneration. This review therefore focuses on the molec- site of injury 1–6 h after muscle damage, followed by
ular mechanisms activated by sex hormones in response to macrophages (*48 h post-injury). Two distinct macro-
skeletal muscle injury due to unaccustomed exercise or phage populations sequentially invade the injured muscle,
trauma. and their transition is important for the regeneration pro-
In this article we review the literature incorporating arti- cess [13, 14]. Phagocytic M1 macrophages secrete pro-
cles from a PubMed search with the keywords ‘androgens’, inflammatory cytokines and promote phagocytosis of
‘estrogens’, ‘phytoestrogens’, ‘estrogen receptor subtype necrotic cell material and satellite cell proliferation,
selective agonists’, ‘SERMS’, ‘SARMS’, ‘skeletal muscle’, whereas the non-phagocytic M2 macrophage population
‘injury’, ‘regeneration’, ‘satellite cells’ and ‘inflammation’. produces anti-inflammatory cytokines, which are vital for
Relevant papers were selected for review, including a few myoblast differentiation and regeneration [13]. Recent data
classical papers on estrogenic and androgenic actions in from Saclier and colleagues confirm that differentially
skeletal muscle. The PubMed search revealed that there are activated, polarized macrophages control myogenic pre-
more ‘early’ (prior to 1965) articles on the effects of cursor fate. In-vitro co-culture experiments of myogenic
androgens on skeletal muscle than there are articles on precursor cells with M1 or M2 macrophages show that M1
estrogens and skeletal muscle. However, the number of macrophages decrease cell fusion, whereas M2 macro-
articles on both androgenic and estrogenic effects on skeletal phages stimulate myotube formation [15]. The authors also
muscle increased sharply in the 1990s, and publication show human data from an eccentric exercise study,
numbers continue to be high to date. In 2012, a total of 188 although with a small sample size, to support the concept
papers were published on the effects of androgens in skeletal that macrophage polarization controls myogenic cell
muscle and 125 articles on estrogens and skeletal muscle. activity. Proliferating myogenin-negative myoblasts were
The high publication numbers reflect the health relevance of detected in regenerating areas containing M1 macrophages
sex hormones and their role in skeletal muscle regeneration. expressing pro-inflammatory markers, whereas myogenin-
We begin this article with a brief general introduction of positive differentiating myogenic precursors were associ-
skeletal muscle regeneration and an overview of the cel- ated with anti-inflammatory M2 macrophages [15]. The
lular mechanisms of estrogenic and androgenic steroids in importance of chemotactic factors, attracting myogenic
skeletal muscle. We assess the latest evidence relating to precursor cells and macrophages to the injury site is
the effects of estrogens and androgens on skeletal muscle revealed in macrophage chemoattractant protein (MCP)-1
Sex Hormones and Skeletal Muscle Regeneration

knockout mice, where regeneration is drastically reduced tissues, such as adipose tissue. Sex steroid hormone targets
[16]. A number of recent in vivo studies delineate some of include the reproductive organs, bone, the vascular,
the mechanisms behind macrophage-mediated muscle immune and nervous system, skeletal muscle and a number
regeneration. In an immunodeficient mouse model, co- of other organ systems [30].
injection of pro-inflammatory macrophages and human Sex steroid hormones can act genomically through ste-
myoblasts increased myoblast proliferation and cell roid receptors with distinct functional domains, including
migration by switching to an anti-inflammatory phenotype transcriptional activation domains (AF-1, AF-2), a DNA-
[17]. Macrophage injection also significantly improves binding domain and a ligand-binding domain. A great
myoblast survival, adhesion and migration in dystrophic number of steroid receptor co-activators have been identi-
mdx mouse muscle [18]. fied, which are recruited into the agonist-bound receptor
There is a tight link between the inflammatory response complex to make DNA accessible and allow the tran-
and activation of myogenic precursor cells, including scriptional process to proceed [31]. In contrast, antagonist-
Pax7? satellite cells. Non-steroidal anti-inflammatory drug bound steroid receptors recruit co-repressors into the
(NSAID) treatment significantly blunts satellite cell acti- receptor complex, which repress transcriptional activation
vation after unaccustomed eccentric exercise [19] or after through activation of histone deacetylases [32]. Over
accustomed endurance exercise [20]. There is evidence that 30,000 androgen receptor (AR) gene targets have been
non-muscle derived cell populations, originating from bone identified in myoblasts in response to androgen treatment
marrow [21] or blood vessels, such as pericytes [18, 22], using ChIP-on-chip technology, including a number of
have the ability to migrate into regenerating skeletal genes involved in muscle growth and development as well
muscle areas and participate in the regeneration process. as micro RNA encoding genes [33]. Estrogen receptors
However, genetic ablation studies have convincingly (ERs) bind to estrogen response elements on target genes
shown that other precursor cells do not compensate suffi- and modulate gene expression by interacting with tran-
ciently for the loss of satellite cells and that muscle scription factors and other co-regulatory factors upon
regeneration is severely compromised in satellite cell ligand binding. Two ERs are known to date (ERa, ERb),
depleted muscle [23–25]. Although satellite cells only with similar amino acid sequences but different tissue
account for approximately 2–5 % of total muscle nuclei, expression levels. It has been shown that ERb reduces
fewer than 10 satellite cells are capable of generating over ERa-regulated gene transcription in bone [34]. Based on
100 new muscle fibres [26]. Satellite cells have the stem these findings, the concept of a ‘Ying Yang’ relationship
cell inherent ability to remain in a quiescent state between between the two receptor isoforms was introduced [34].
the sarcolemma and basal lamina. Maintenance of the There is evidence showing that heterodimerization occurs
quiescent state was recently shown to be regulated by not only between ERa and ERb but also between ERa and
microRNA-489, which post-transcriptionally suppresses an AR, adding additional complexity to sex steroid hormone
oncogene product involved in the proliferative expansion signalling. Co-expression of ER and AR in the presence of
of satellite cells [27]. Satellite cells are activated in 17b-estradiol (E2) dose-dependently decreases AR tran-
response to a myotrauma and begin to proliferate with a scriptional activity [35]. Both ERa and ERb are expressed
doubling time of approximately 17 h in the mouse [28]. in skeletal muscle of mice [36, 37], rats [38, 39] and
Satellite cells proliferate for 2–3 days and then withdraw humans [40–42].
from the cell cycle to differentiate or return to quiescence In addition to the classical transcriptional pathway
(asymmetric division). Differentiating satellite cells are mechanism (steroid hormones ? steroid recep-
Pax7, MyoD and Myf5 positive, whereas satellite cells tors ? hormone response element [HRE]-mediated gene
returning to quiescence are only Pax7 positive [18]. induction) estrogens and androgens are also able to induce
Regenerating fibres can be detected by central nucleation, a rapid (seconds to minutes), HRE-independent effects
sign for satellite cells fusing with existing fibres or aligning through protein–protein interactions, or non-genomic/non-
to form new myofibres to contribute with their DNA to transcriptional effects through intracellular kinase signal-
muscle growth and regeneration. Depending on the type ling pathways, by impacting on membrane fluidity or by
and severity of the injury, muscle regeneration can be signalling through plasma membrane-bound steroid
complete within 7–21 days post-injury [29]. receptors or non-nuclear receptors [43, 44]. There are
several lines of evidence providing compelling evidence
1.2 Cellular Mechanisms of Estrogenic for the existence of non-transcriptional estrogen and
and Androgenic Hormones androgen pathways. For example, pituitary cell prolactin
secretion is stimulated only 5 min after testosterone
Estrogens, progestins and androgens are sex steroids pro- administration [45]. Also, estrogen induces rapid vasodi-
duced by the reproductive organs and ‘non-classical’ lation in vascular walls [46] and neuronal excitability in the
M. Velders, P. Diel

nervous system [47]. A number of studies have provided muscle membranes from exercise-induced muscle damage.
evidence for the existence of membrane-associated estro- Since then it has been argued that CK is a poor marker of
gen-binding sites, including the pioneering work of Pietras muscle damage, due to high intra-individual variability as
and Szego [48], which identified ER cell membrane sub- well as the dynamic nature of CK release and clearance
fractions in isolated endometrial cells. There is now gen- [58]. However, when systemic effects are removed and
eral agreement that ERs can localize to the cell membrane skeletal muscle is studied in isolation in vitro, studies have
and to membrane-associated structures such as caveolae shown that basal and contraction-induced CK release was
[49], but other non-steroid membrane-associated receptors still greater in male and OVX female rats than in intact
may also interact with estrogens to activate downstream female and estrogen-treated male rat muscle [59, 60].
second messengers and protein kinases [50]. Findings from our laboratory agree with other published
Non-transcriptional steroid hormone signalling mecha- data from rodent studies [55, 56, 61] by showing that
nisms can be studied using cells expressing mutated steroid elimination of ovarian hormone production by OVX or
receptors, steroid receptor blockers, inhibitors of RNA or selective knockout of the ERb isoform results in higher
protein synthesis or steroid hormones linked to cell- serum CK activity levels after muscle injury than in intact
impermeable molecules such as bovine serum albumin rats [3] Furthermore, treating OVX rats with the natural
[50]. Studies using the AR-negative L6 myoblast cell line estrogenic ligand E2 and other substances with affinity to
or bovine serum albumin-linked testosterone have provided the ERs (ERa and ERb selective agonists and the phyto-
evidence for the existence of non-transcriptional androgen estrogen genistein) attenuated serum CK activity after
pathways by showing that testosterone treatment stimulates toxin-induced muscle damage [3].
myoblast proliferation and differentiation via G-protein These findings argue in favor of estrogen’s non-tran-
coupled signalling pathways [51]. In isolated skeletal scriptional effects, acting as a strong antioxidant and
muscle, G-protein inhibitors blocked the fast (\2 min), membrane stabilizer and protecting sarcolemmal integrity
transient increase in intracellular calcium in response to [3, 62]. While there is still debate on the role of estrogens
testosterone treatment [52]. This non-transcriptional tes- in preventing primary, structural muscle damage, particu-
tosterone-induced mechanism in skeletal muscle may be larly in human exercise studies, evidence is more conclu-
relevant for the transcriptional regulation of calcium-sen- sive for the role of estrogens in preventing secondary
sitive transcription factors, such as nuclear factor of acti- muscle damage by modulating inflammatory dynamics of
vated T-cells (NFAT) or genes containing calcium- skeletal muscle after eccentric exercise [4, 61], notexin
sensitive promoter regions (e.g. calcium response element injury [3], ischaemia/reperfusion injury [63] or recovery
[CARE]) [53]. Additional evidence for AR independent after disuse [64, 65]. The inflammatory cascade initiated
androgen actions comes from studies using AR blockers. after injury or unaccustomed muscle use involves an early
Dihydrotestosterone (DHT) treatment of isolated mouse invasion of neutrophils followed by the infiltration of M1
muscle fibres, in combination with AR blockers, but not in and M2 macrophages, which secrete pro- and anti-inflam-
the presence of epidermal growth factor (EGF) receptor matory cytokines, respectively [14, 66, 67]. Neutrophil
inhibitors, increases isometric force in fast muscle fibres by invasion is exacerbated by OVX [64] and attenuated by
activating the mitogen-activated protein kinase (MAPK) estrogen treatment [3, 5]. While neutrophils serve impor-
pathway [54]. tant functions clearing tissue debris, they lack the ability to
differentiate between intact and damaged proteins [68]. A
study using an ER antagonist has shown that the attenu-
2 Sex Steroids Augment Skeletal Muscle Regeneration: ating effects of estrogens on leukocyte infiltration is a non-
Animal Studies ER mediated process [4] and is most likely due to the
positive effects of estrogens on lipid membrane fluidity
2.1 Transcriptional and Non-Transcriptional Effects [69]. Another published E2-mediated non-transcriptional
of Estrogens During Muscle Regeneration mechanism relates to the ability of E2 to reduce inflam-
matory gene expression in macrophages by inhibiting
Findings from Amelink, Bär and colleagues in the late nuclear factor kappa-light-chain-enhancer of activated B
1980s and 1990s first showed that serum activities of cells (NF-jB) intracellular transport [70]. Decreased NF-
muscle enzymes, such as creatine kinase (CK), were ele- jB-mediated gene expression may partially explain our
vated in male rats after muscle-damaging exercise com- findings of reduced gene expression levels of the pro-
pared with gonad-intact female rats, while ovariectomized inflammatory cytokine tumor necrosis factor (TNF)-a and
(OVX) female rats showed CK elevations similar to those increased expression of the anti-inflammatory cytokine
of male rats [55–57]. These findings first suggested that interleukin (IL)-10 in skeletal muscle of estrogen-treated
estrogens may play an important role in protecting skeletal OVX rats [3].
Sex Hormones and Skeletal Muscle Regeneration

While excessive skeletal muscle inflammation leads to estrogens on inflammatory dynamics during muscle
tissue injury by oxidative damage [71], a certain level of regeneration. However, there is limited evidence to suggest
inflammation is required to initiate the muscle-regeneration that the effects of androgens on inflammatory cytokine
process by clearing cellular debris and by stimulating profiles are age dependent. Nandrolone treatment attenu-
satellite cell function. Satellite cell activation, proliferation ates overload-induced IL-1b expression only in old rodent
and fusion with existing myofibres are vital processes muscle and IL-6 in young and old muscle [81].
during skeletal muscle regeneration [23]. Studies using
OVX rats as a model of menopause have shown that
satellite cell activation is reduced after toxin injury [3] or 3 Effects of Hormonal Status on Muscle Damage,
eccentric exercise [4, 5, 72], while estrogen substitution Repair and Function: Human Studies
leads to an augmented satellite cell response. The stimu-
lating effects of estrogens on satellite cell activity are also 3.1 Effects of Estrogenic Status on Human Skeletal
evident when male rats are treated with E2 before eccentric Muscle Health
exercise [73]. The effects of E2 appear to be ER mediated
[4] and the absence of the ERb receptor isoform in While animal studies have convincingly shown that estro-
knockout mice particularly impacts negatively on markers gens ameliorate post-exercise muscle damage and inflam-
of satellite cell activation, such as Pax7 or MyoD [3]. mation [4] and enhance skeletal muscle repair processes
While our data suggest that the ERb receptor isoform is the [3–5], evidence from human studies is less abundant. An
predominant ER isoform inducing E2-mediated transcrip- excellent point/counterpoint discussion on the role of
tional responses during muscle regeneration, another study estrogens and sex on post-exercise indices of muscle
using an exercise-induced injury model has shown that the damage, inflammation and repair has highlighted the fact
ERa selective ligand propylpyrazole triol (PPT), similar to that there is a discrepancy between findings from animal
E2, augmented satellite cell number and activation [74]. versus human studies relating to the effects of estrogens on
muscle damage, inflammation and repair [62, 82, 83]. This
2.2 Androgens Promote the Skeletal Muscle may be largely because most studies have investigated sex
Regeneration Process differences rather than studying human populations dis-
cordant for estrogen levels. There are a number of sex-
The effects of anabolic steroids on muscle anabolism and based differences other than estrogen status, which may
regeneration were first studied using rodents in the 1960s affect outcome measures. A more promising approach for
and 1970s [75, 76]. Testosterone supplementation of cas- studying the specific role of estrogens on skeletal muscle in
trated young and aged mice increases the number of pro- humans, as suggested by Phillips in a point/counterpoint
liferating satellite cells and fibre cross-sectional area in discussion by Pizza et al. [83] is a comparison between
regenerating muscle [77]. Administration of nandrolone to people of the same sex discordant for estrogen status, such
mice after toxin-induced muscle injury augments growth- as women before and after surgical ovariectomy, HRT-
related gene expression, muscle weight and size of regen- treated versus untreated post-menopausal women, or men
erating fibres [78]. The effects of nandrolone appear to be treated with high dosages of estrogen after a sex change. A
particularly beneficial to a slow fibre-type muscle (e.g. study comparing twins discordant for HRT use showed that
soleus) in terms of muscle mass and myosin heavy chain the twins using HRT exhibited significantly greater muscle
expression [6]. However, fast fibre type muscles (extensor strength and mass than the twins not using HRT [84]. A
digitorum longus [EDL]) also respond to nandrolone year-long HRT plus exercise intervention with post-men-
treatment after toxin injury with an increased frequency of opausal women showed that exercise and HRT had addi-
large fibres and enhanced expression of MyoD [78]. While tive positive effects on functional measures compared with
nandrolone treatment before surgical ablation of the gas- HRT alone [85]. More recent evidence suggests that
trocnemius and soleus muscles did not enhance compen- estrogen treatment is not only beneficial for skeletal muscle
satory hypertrophy of the plantaris muscle, steroid homeostasis and regeneration in older women, but also
treatment significantly reduced the mean size of the attenuates neutrophil infiltration after damaging eccentric
necrotic area [79]. muscle contractions in young men [86].
There appears to be a common mechanistic basis for the Dieli-Conwright and colleagues [87–91] have provided
effects of androgens and estrogens on muscle-regeneration mechanistic evidence for the beneficial effects of HRT on
processes. Both steroid nuclear receptor classes promote human skeletal muscle mass and function. They showed
satellite cell activation and proliferation and stimulate that expression of genes promoting muscle growth (myo-
expression of growth-promoting genes [3, 5, 78, 80]. Less genic regulatory factors) was enhanced, while expression
is known about the effects of androgens compared with of negative regulators of muscle growth, such as myostatin
M. Velders, P. Diel

or proteolytic factors, was reduced in skeletal muscle of the potential to promote skeletal muscle regenerative pro-
post-menopausal women using HRT compared with post- cesses based on their ability to promote protein synthesis
menopausal women not using HRT [87, 88]. The reduction and satellite cell activation [6, 80, 100]. Supraphysiological
of myostatin and activin IIb receptor expression was also doses of testosterone have been shown to increase satellite
greater after acute eccentric exercise in women using HRT, cell numbers, and this change was significantly correlated
which may be one of the molecular mechanisms underlying with total and free testosterone concentrations [101]. In
the effects of estrogens on muscle mass and strength addition, testosterone also modulates cellular protein deg-
preservation [87]. Also, ER transcriptional activity, deter- radation and inflammatory processes by reducing the
mined via messenger RNA (mRNA) expression of ER co- expression of the ubiquitin ligase muscle atrophy F-box
regulators, was significantly greater following eccentric (MAFbx) [102] and by stimulating the expression of anti-
exercise in skeletal muscle of women using HRT versus inflammatory cytokines [103].
post-menopausal women not using HRT [90]. The study is Although there is a clear sex difference in endogenous
the first to show that acute eccentric exercise enhances ER testosterone, with levels in women being one order of
transcriptional activity and that ER co-regulator gene magnitude lower than in men, total serum testosterone
expression is further enhanced in postmenopausal women levels are significantly reduced in late post-menopausal
using HRT. The downstream signalling pathways are women, and estrogen therapies lead to further reductions in
starting to be deciphered, and accumulating evidence testosterone levels [104]. The physiological significance of
suggests that the positive effects of HRT on muscle mass reduced testosterone levels for muscle mass maintenance
and function observed in human studies are due to estro- or muscle regenerative capacity in response to estrogen
gens creating a pro-anabolic skeletal muscle environment therapies requires further study. Although androgen thera-
[68, 92]. pies in women remain highly controversial, older females
Evidence from rodent studies supports the claim that benefit from androgen treatment in terms of muscle protein
there is an inverse relationship between protective effects synthesis [105].
of HRT and the duration of hypoestrogenicity [93]. This Circulating anabolic hormone levels such as testosterone
‘window of opportunity’ hypothesis is also supported by are significantly elevated in response to high volume,
human studies showing that cardiovascular benefits are moderate- to high-intensity exercise using large muscle
greatest and risks are minimized when HRT is initiated groups [106]. The hormonal response to strength training,
before 60 years of age or within 10 years after the onset of consisting of elevated free testosterone and insulin-like
menopause [94]. Gene expression profiling of skeletal growth factor IGF-1 levels at rest and during exercise, can
muscle from early-stage post-menopausal women before also be observed in older men, although at lower absolute
and 12 months after an intervention, consisting of a plyo- levels than in younger men [106]. Recent evidence sug-
metric power training and HRT, revealed significantly gests that oral dehydroepiandrosterone (DHEA) supple-
enhanced gene expression related to mitochondria func- mentation protects skeletal muscle of young men from
tionality, carbohydrate metabolism and calcium signalling exercise-induced damage [107].
[92]. More research is needed to determine the effects of
HRT initiation from the onset of menopause on skeletal
muscle health. 4 Sex Hormones: Therapy Implications

3.2 Androgen Levels in Humans Impact on Muscle The importance of female sex hormones as major deter-
Mass and Strength minants of movement drive and physical activity levels has
been clearly demonstrated in rodent studies showing that
Andropause in men is associated with physical and emo- (i) female rodents are generally more active than male
tional changes caused by decreasing androgen levels. Cir- rodents [108, 109], (ii) gonadectomized male and female
culating testosterone levels decline 1–3 % per year starting rodents exhibit reduced levels of voluntary physical
from the fourth decade of life [95]. Men are generally activity [110], and (iii) treating gonadecomized male
considered hypogonadal when morning serum total tes- rodents with testosterone or E2 and female rats with E2
tosterone falls below 300–500 ng/dL [96]. ARs in human significantly increases voluntary physical activity. While a
skeletal muscle mediate the effects of androgens on myo- causative effect between sex hormones and physical
nuclear accretion and protein synthesis [51, 80, 97], and activity drive has yet to be shown in human studies [111],
several studies have shown that testosterone administration the positive effects of both male and female sex hormones
improves muscle mass and strength in hypogonadal men on muscle mass and strength, as well as attenuation of
[96], but also in young eugonadal men [98] and in men muscle damage after eccentric exercise are well docu-
with HIV-induced muscle wasting [99]. Androgens have mented [84, 112–114].
Sex Hormones and Skeletal Muscle Regeneration

4.1 Selective Estrogen and Androgen Receptor the regulation of muscle growth and regeneration in post-
Modulators as Therapeutic Agents menopausal women.
There are reports on the abuse potential of SARMs for
Selective estrogen and androgen receptor modulators doping purposes in sports [122]. However, more studies are
(SERMs and SARMs) exhibit tissue-selective estrogenic or needed in patient populations to examine the effects of
androgenic activity. They are developed to exploit the exercise on SARM and SERM metabolism and the impli-
beneficial effects of estrogens and androgens on various cations for signalling events in muscle cells.
target tissues such as skeletal muscle, bone and the vas-
cular system without the known estrogen-related adverse 4.2 Estrogen Receptor Subtype Selective Agonists
effects on the breast and prostate.
Data from our laboratory have shown that the prohor- Estrogen-mediated cellular proliferation is primarily
mone 19-norandrostenedione possesses SARM-like prop- transmitted through the ERa, which is also upregulated in
erties by selectively stimulating skeletal muscle growth human breast cancer tissue [123]. Activation of the ERb
without affecting prostate proliferation [115]. Similar isoforms, on the other hand, reduces inflammation in
results were obtained by Gao and colleagues, showing that chronic inflammatory diseases and ERb-selective ligands
the SARM S-4 induces strong anabolic effects in skeletal have the benefit of being non-uterotrophic and non-mam-
muscle, bone and the pituitary gland of orchiectomized motrophic [124].
rats without causing adverse effects on the prostate A number of studies from our laboratory using a rat
compared with DHT [116]. In addition to counteracting model of menopause (OVX) have shown that ER subtype
castration-induced skeletal muscle atrophy, SARMs have selective ligands induce protective effects in skeletal
also been reported to attenuate glucocorticoid-induced muscle [3, 125, 126], intestinal cells [127] and bone [128].
atrophy by reducing the expression of ubiquitin ligases By using both ER knockout mice and ER subtype selective
and by inhibiting dephosphorylation of factors involved in agonists, we found that the ERb isoform is primarily
protein synthesis [117]. A number of SARMs are cur- responsible for inducing protective effects in skeletal
rently being tested in clinical trials as function-promoting muscle after injury [3], in response to high-intensity
therapies, including GTx-024 (enobosarm), where treat- treadmill running exercise [125] or during nutrition-
ment dose-dependently increases lean body mass and induced obesity [126]. Our data also suggest that the ERb
physical function in elderly men and post-menopausal isoform activates an anabolic transcriptional programme,
women [118]. stimulating muscle hypertrophy in orchiectomized male
SERMs have been in clinical use for over 50 years rats and OVX female rats [3, 126]. While another study has
[119], and large clinical trials such as the STAR (Study of also found beneficial effects of an ERa-selective ligand on
Tamoxifen and Raloxifene) trial by the US National Can- satellite cell activation after exercise-induced muscle injury
cer Institute have investigated the effects of SERMs on [74], there is accumulating evidence in favor of ERb
reducing the incidence of breast cancer in 19,737 post- subtype selective agonists as therapeutic agents to reduce
menopausal women [120]. Research is therefore warranted inflammation in chronic inflammatory diseases [129, 130]
to examine the effects of these ligands on skeletal muscle or to protect the intestinal tract from tumour development
health and function in post-menopausal women who are at by inducing anti-proliferative and pro-apoptotic effects
risk of developing sarcopenia. To date there are limited [127]. There are efforts underway to develop both steroidal
data on the effects of SERMs on skeletal muscle adaptation and non-steroidal ERb-selective agonists to treat inflam-
or regeneration in humans. Subcutaneous tamoxifen treat- matory diseases [131].
ment induced estrogen-like protective effects on skeletal Soy isoflavones display estrogenic activity based on
muscle membrane integrity after electrical stimulation in structural similarities to the natural ligand E2 and are
male and female rats [121]. Isolated human skeletal muscle therefore referred to as phytoestrogens. The non-steroidal
cells in culture respond to E2, tamoxifen and raloxifene phytoestrogen genistein binds with higher affinity to the
treatment with increased ERa and steroid receptor coacti- ERb compared with the ERa [132] and has been shown to
vator (SRC) and decreased silencing mediator for retinoid stimulate the expression of satellite cell markers in injured
and thyroid hormone receptors (SMRT) mRNA expression skeletal muscle [3]. In addition, genistein has recently been
[91]. MyoD mRNA expression decreased in response to shown to act as an AR modulator. In AR reporter mice,
both tamoxifen and raloxifene, and glucose transporter genistein was anti-androgenic in the prostate, testis and
(GLUT)-4 expression increased with raloxifene and brain of intact mice and pro-androgenic in the prostate and
decreased with tamoxifen [91]. These findings suggest that brain of castrated mice [133]. The effects of isoflavones on
SERMs have differential effects on myogenic gene skeletal muscle growth appear to be concentration depen-
expression pattern and these changes may be important for dent. Genistein affects cell growth by inhibiting protein
M. Velders, P. Diel

tyrosine kinases, such as the IGF-1 receptor. Cell culture Studies have shown that estrogens and androgens impact
studies have shown that genistein has no effect on IGF-1 positively on satellite cell activation and proliferation
and IGF-1 receptor expression at dietary concentrations up in vivo [3–5, 101]. Aging muscle maintains the ability to
to 10 lmol/L [134], whereas proliferation rates of porcine recruit satellite cells in response to strength training [8].
satellite cells are reduced when treated with genistein Therefore, future studies should further investigate the
concentrations [1 lmol/L [135]. In addition, genistein effects of combined hormonal and exercise interventions
affects muscle cell protein metabolism in a dose-dependent on muscle health. Studies are also required to study the
manner, reducing protein synthesis at higher concentrations effects of hormone therapy timing on muscle regeneration
(100 lmol/L genistein) and reducing protein degradation at and inflammation following exercise or disuse in older
lower concentrations (0.1 lmol/L genistein) [136]. One people. Exercise science can contribute to this field by
human study comparing the effects of 28 days of daily soy investigating the effects of specific exercise regimens in
and dairy milk ingestion on skeletal muscle inflammation combination with hormonal therapies on estrogen- and
in post-menopausal women after muscle damaging down- androgen-regulated molecular responses in skeletal muscle.
hill running exercise showed no significant effects of soy
ingestion on expression of inflammatory or proteolytic Acknowledgments The authors have no conflicts of interest rele-
vant to this review to declare. No specific funding sources were used
genes [137]. The dose-dependent effects of isoflavones on to write this manuscript.
human skeletal muscle myogenic responses after exercise
remain to be determined.
References

5 Conclusion and Outlook 1. Dreyer HC, Blanco CE, Sattler FR, et al. Satellite cell numbers
in young and older men 24 hours after eccentric exercise.
Muscle Nerve. 2006;33(2):242–53. doi:10.1002/mus.20461.
Aging is associated with a natural decline in sex hormone 2. Carlson BM, Faulkner JA. Muscle transplantation between
levels in both men and women. Loss of muscle mass with young and old rats: age of host determines recovery. Am J
aging results in functional impairment and loss of inde- Physiol. 1989;256(6 Pt 1):C1262-6.
pendence. Implications of the age-related hormonal 3. Velders M, Schleipen B, Fritzemeier KH, et al. Selective
estrogen receptor-beta activation stimulates skeletal muscle
decline for skeletal muscle ultrastructural damage and growth and regeneration. FASEB J. 2012;26(5):1909–20.
regeneration has been more clearly established in animal doi:10.1096/fj.11-194779.
than in human studies [3, 5, 138]. Inconsistent results 4. Enns DL, Iqbal S, Tiidus PM. Oestrogen receptors mediate
reported from human studies are ascribed to heteroge- oestrogen-induced increases in post-exercise rat skeletal muscle
satellite cells. Acta Physiol (Oxf). 2008;194(1):81–93.
neous study populations and general sex-based differences 5. Enns DL, Tiidus PM. Estrogen influences satellite cell activation
other than sex hormone status [83]. Despite these dis- and proliferation following downhill running in rats. J Appl
crepancies, there is convincing evidence in favour of sex Physiol. 2008;104(2):347–53.
hormones promoting skeletal muscle homeostasis and 6. Ferry A, Noirez P, Page CL, et al. Effects of anabolic/andro-
genic steroids on regenerating skeletal muscles in the rat. Acta
regeneration. Androgens are used as therapeutic agents to Physiol Scand. 1999;166(2):105–10.
reduce degeneration and enhance protein synthesis in 7. Bhasin S, Woodhouse L, Storer TW. Proof of the effect of
muscle dystrophy conditions and other wasting diseases, testosterone on skeletal muscle. J Endocrinol. 2001;170(1):
such as HIV [99, 139]. Likewise, HRT creates a pro- 27–38. pii: JOE04157.
8. Roth SM, Martel GF, Ivey FM, et al. Skeletal muscle satellite
anabolic skeletal muscle environment in post-menopausal cell characteristics in young and older men and women after
women [68, 88]. Although some of underlying mecha- heavy resistance strength training. J Gerontol Ser A Biol Sci
nisms responsible for the anabolic effects of HRT have Med Sci. 2001;56(6):B240–7.
been uncovered [88, 90], more work is necessary to 9. Carlson BM, Gutmann E. Development of contractile properties
of minced muscle regenerates in the rat. Exp Neurol.
determine the effects of sex hormones on the expression 1972;36(2):239–49. pii: 0014-4886(72)90020-9.
and activity of skeletal muscle myogenic and atrogenic 10. Turner NJ, Badylak SF. Regeneration of skeletal muscle. Cell
signalling molecules, in order to identify potential targets Tissue Res. 2012;347(3):759–74. doi:10.1007/s00441-011-
for pharmacological interventions [140]. Since a classical 1185-7.
11. Belcastro AN, Shewchuk LD, Raj DA. Exercise-induced muscle
estrogen-containing HRT has been associated with an injury: a calpain hypothesis. Mol Cell Biochem. 1998;179(1–2):
increased breast cancer risk [141], ER subtype selective 135–45.
ligands and tissue selective receptor modulators (SERMs, 12. Wehling M, Spencer MJ, Tidball JG. A nitric oxide synthase
SARMs) are developed to reduce proliferation in mitotic transgene ameliorates muscular dystrophy in mdx mice. J Cell
Biol. 2001;155(1):123–31. doi:10.1083/jcb.200105110155/1/123.
tissues, such as the mammary gland and prostate, while 13. Tidball JG, Villalta SA. Regulatory interactions between muscle
maintaining the estrogenic and androgenic benefits in and the immune system during muscle regeneration. Am J
skeletal muscle and bone. Physiol Regul Integr Comp Physiol. 2010;298(5):R1173–87.
Sex Hormones and Skeletal Muscle Regeneration

14. Tidball JG. Inflammatory processes in muscle injury and repair. roles in muscle development and function. Mol Endocrinol.
Am J Physiol Regul Integr Comp Physiol. 2005;288(2): 2010;24(8):1665–74. doi:10.1210/me.2010-0138.
R345–53. 34. Lindberg MK, Moverare S, Skrtic S, et al. Estrogen receptor
15. Saclier M, Yacoub-Youssef H, Mackey AL, et al. Differentially (ER)-beta reduces ERalpha-regulated gene transcription, sup-
activated macrophages orchestrate myogenic precursor cell fate porting a ‘‘ying yang’’ relationship between ERalpha and
during human skeletal muscle regeneration. Stem Cells. 2012. ERbeta in mice. Mol Endocrinol. 2003;17(2):203–8.
doi:10.1002/stem.1288. 35. Kumar MV, Leo ME, Tindall DJ. Modulation of androgen
16. Shireman PK, Contreras-Shannon V, Ochoa O, et al. MCP-1 receptor transcriptional activity by the estrogen receptor. J An-
deficiency causes altered inflammation with impaired skeletal drol. 1994;15(6):534–42.
muscle regeneration. J Leukoc Biol. 2007;81(3):775–85. 36. Milanesi L, Russo de Boland A, Boland R. Expression and
17. Bencze M, Negroni E, Vallese D, et al. Proinflammatory mac- localization of estrogen receptor alpha in the C2C12 murine
rophages enhance the regenerative capacity of human myoblasts skeletal muscle cell line. J Cell Biochem. 2008;104(4):1254–73.
by modifying their kinetics of proliferation and differentiation. 37. Milanesi L, Vasconsuelo A, de Boland AR, et al. Expression and
Mol Ther. 2012;20(11):2168–79. doi:10.1038/mt.2012.189. subcellular distribution of native estrogen receptor beta in
18. Tedesco FS, Dellavalle A, Diaz-Manera J, et al. Repairing murine C2C12 cells and skeletal muscle tissue. Steroids.
skeletal muscle: regenerative potential of skeletal muscle stem 2009;74(6):489–97.
cells. J Clin Invest. 2010;120(1):11–9. doi:10.1172/JCI40373. 38. Lemoine S, Granier P, Tiffoche C, et al. Effect of endurance
19. Mikkelsen UR, Langberg H, Helmark IC, et al. Local NSAID training on oestrogen receptor alpha transcripts in rat skeletal
infusion inhibits satellite cell proliferation in human skeletal muscle. Acta Physiol Scand. 2002;174(3):283–9.
muscle after eccentric exercise. J Appl Physiol. 2009;107(5): 39. Lemoine S, Granier P, Tiffoche C, et al. Effect of endurance
1600–11. training on oestrogen receptor alpha expression in different rat
20. Mackey AL, Kjaer M, Dandanell S, et al. The influence of anti- skeletal muscle type. Acta Physiol Scand. 2002;175(3):211–7.
inflammatory medication on exercise-induced myogenic pre- 40. Wiik A, Glenmark B, Ekman M, et al. Oestrogen receptor beta is
cursor cell responses in humans. J Appl Physiol. expressed in adult human skeletal muscle both at the mRNA and
2007;103(2):425–31. doi:10.1152/japplphysiol.00157.2007. protein level. Acta Physiol Scand. 2003;179(4):381–7.
21. Ferrari G, Cusella-De Angelis G, Coletta M, et al. Muscle 41. Wiik A, Ekman M, Morgan G, et al. Oestrogen receptor beta is
regeneration by bone marrow-derived myogenic progenitors. present in both muscle fibres and endothelial cells within human
Science. 1998;279(5356):1528–30. skeletal muscle tissue. Histochem Cell Biol. 2005;124(2):161–5.
22. Dellavalle A, Maroli G, Covarello D, et al. Pericytes resident in 42. Wiik A, Ekman M, Johansson O, et al. Expression of both
postnatal skeletal muscle differentiate into muscle fibres and oestrogen receptor alpha and beta in human skeletal muscle
generate satellite cells. Nat Commun. 2011;2:499. doi:10.1038/ tissue. Histochem Cell Biol. 2009;131(2):181–9.
ncomms1508. 43. Hall JM, Couse JF, Korach KS. The multifaceted mechanisms of
23. Sambasivan R, Yao R, Kissenpfennig A, et al. Pax7-expressing estradiol and estrogen receptor signaling. J Biol Chem.
satellite cells are indispensable for adult skeletal muscle 2001;276(40):36869–72. doi:10.1074/jbc.R100029200R1000
regeneration. Development. 2011;138(17):3647–56. doi:10. 29200.
1242/dev.067587. 44. Dubois V, Laurent M, Boonen S, et al. Androgens and skeletal
24. Lepper C, Partridge TA, Fan CM. An absolute requirement for muscle: cellular and molecular action mechanisms underlying
Pax7-positive satellite cells in acute injury-induced skeletal the anabolic actions. Cell Mol Life Sci. 2012;69(10):1651–67.
muscle regeneration. Development. 2011;138(17):3639–46. doi:10.1007/s00018-011-0883-3.
doi:10.1242/dev.067595. 45. Christian HC, Rolls NJ, Morris JF. Nongenomic actions of
25. Kuang S, Charge SB, Seale P, et al. Distinct roles for Pax7 and testosterone on a subset of lactotrophs in the male rat pituitary.
Pax3 in adult regenerative myogenesis. J Cell Biol. Endocrinology. 2000;141(9):3111–9.
2006;172(1):103–13. 46. Karas RH, Hodgin JB, Kwoun M, et al. Estrogen inhibits the
26. Collins CA, Olsen I, Zammit PS, et al. Stem cell function, self- vascular injury response in estrogen receptor beta-deficient
renewal, and behavioral heterogeneity of cells from the adult female mice. Proc Natl Acad Sci USA. 1999;96(26):15133–6.
muscle satellite cell niche. Cell. 2005;122(2):289–301. doi:10. 47. McEwen BS, Alves SE. Estrogen actions in the central nervous
1016/j.cell.2005.05.010. system. Endocr Rev. 1999;20(3):279–307.
27. Cheung TH, Quach NL, Charville GW, et al. Maintenance of 48. Pietras RJ, Szego CM. Specific binding sites for oestrogen at the
muscle stem-cell quiescence by microRNA-489. Nature. outer surfaces of isolated endometrial cells. Nature.
2012;482(7386):524–8. doi:10.1038/nature10834. 1977;265(5589):69–72.
28. Zammit PS, Heslop L, Hudon V, et al. Kinetics of myoblast 49. Chambliss KL, Yuhanna IS, Anderson RG, et al. ERbeta has
proliferation show that resident satellite cells are competent to nongenomic action in caveolae. Mol Endocrinol. 2002;16(5):
fully regenerate skeletal muscle fibers. Exp Cell Res. 938–46.
2002;281(1):39–49. pii: S0014482702956533. 50. Simoncini T, Genazzani AR. Non-genomic actions of sex ste-
29. Hawke TJ, Garry DJ. Myogenic satellite cells: physiology to roid hormones. Eur J Endocrinol. 2003;148(3):281–92.
molecular biology. J Appl Physiol. 2001;91(2):534–51. 51. Fu R, Liu J, Fan J, et al. Novel evidence that testosterone pro-
30. Wierman ME. Sex steroid effects at target tissues: mechanisms motes cell proliferation and differentiation via G protein-cou-
of action. Adv Physiol Educ. 2007;31(1):26–33. doi:10.1152/ pled receptors in the rat L6 skeletal muscle myoblast cell line.
advan.00086.2006. J Cell Physiol. 2012;227(1):98–107. doi:10.1002/jcp.22710.
31. Beato M, Klug J. Steroid hormone receptors: an update. Hum 52. Estrada M, Espinosa A, Muller M, et al. Testosterone stimulates
Reprod Update. 2000;6(3):225–36. intracellular calcium release and mitogen-activated protein
32. Wierman ME, Kohrt WM. Vascular and metabolic effects of sex kinases via a G protein-coupled receptor in skeletal muscle cells.
steroids: new insights into clinical trials. Reprod Sci. Endocrinology. 2003;144(8):3586–97.
2007;14(4):300–14. doi:10.1177/1933719107303673. 53. Rahman F, Christian HC. Non-classical actions of testosterone:
33. Wyce A, Bai Y, Nagpal S, et al. Research resource: the an update. Trends Endocrinol Metab. 2007;18(10):371–8.
androgen receptor modulates expression of genes with critical doi:10.1016/j.tem.2007.09.004.
M. Velders, P. Diel

54. Hamdi MM, Mutungi G. Dihydrotestosterone activates the 75. Koller M. Weight changes in the anterior tibial muscle of the
MAPK pathway and modulates maximum isometric force prepuberal rat in normal conditions, after castration and after
through the EGF receptor in isolated intact mouse skeletal treatment with 2 anabolic steroids. Boll Soc Ital Biol Sper.
muscle fibres. J Physiol. 2010;588(Pt 3):511–25. doi:10.1113/ 1960;36:756–9.
jphysiol.2009.182162. 76. Kuc A, Poplawski W, Targonski R, et al. Influence of nerobolil
55. Bär PR, Amelink GJ, Oldenburg B, et al. Prevention of exercise- on regeneration and histochemical reactions in striated muscles.
induced muscle membrane damage by oestradiol. Life Sci. Folia Morphol (Warsz). 1973;32(1):89–101.
1988;42(26):2677–81. 77. Serra C, Tangherlini F, Rudy S, et al. Testosterone improves the
56. Amelink GJ, Bär PR. Exercise-induced muscle protein leakage regeneration of old and young mouse skeletal muscle. J Gerontol
in the rat: effects of hormonal manipulation. J Neurol Sci. Ser A Biol Sci Med Sci. 2013;68(1):17–26. doi:10.1093/gerona/
1986;76(1):61–8. gls083.
57. Bär PR, Amelink GJ. Protection against muscle damage exerted 78. Wagner Alves de Souza R, Goncalves W, Garrido Cavalcante
by oestrogen: hormonal or antioxidant action? Biochem Soc WL, et al. Nandrolone stimulates MyoD expression during
Trans. 1997;25(1):50–4. muscle regeneration in the condition of myonecrosis induced by
58. Friden J, Lieber RL. Serum creatine kinase level is a poor Bothrops jararacussu venom poisoning. J Toxicol Environ
predictor of muscle function after injury. Scand J Med Sci Health A. 2010;73(13–14):934–43. doi:10.1080/152873910037
Sports. 2001;11(2):126–7. 51729.
59. Amelink GJ, Koot RW, Erich WB, et al. Sex-linked variation in 79. Tamaki T, Uchiyama Y, Okada Y, et al. Anabolic-androgenic
creatine kinase release, and its dependence on oestradiol, can be steroid does not enhance compensatory muscle hypertrophy but
demonstrated in an in-vitro rat skeletal muscle preparation. Acta significantly diminish muscle damages in the rat surgical abla-
Physiol Scand. 1990;138(2):115–24. tion model. Histochem Cell Biol. 2009;132(1):71–81. doi:10.
60. Persky AM, Green PS, Stubley L, et al. Protective effect of 1007/s00418-009-0584-2.
estrogens against oxidative damage to heart and skeletal muscle 80. Chen Y, Zajac JD, MacLean HE. Androgen regulation of
in vivo and in vitro. Proc Soc Exp Biol Med. 2000;223(1): satellite cell function. J Endocrinol. 2005;186(1):21–31.
59–66. 81. Thompson RW, McClung JM, Baltgalvis KA, et al. Modulation
61. Tiidus PM, Holden D, Bombardier E, et al. Estrogen effect on of overload-induced inflammation by aging and anabolic steroid
post-exercise skeletal muscle neutrophil infiltration and calpain administration. Exp Gerontol. 2006;41(11):1136–48. doi:10.
activity. Can J Physiol Pharmacol. 2001;79(5):400–6. 1016/j.exger.2006.08.013.
62. Tiidus PM. Can oestrogen influence skeletal muscle damage, 82. Tiidus PM, Enns DL. Point:counterpoint: estrogen and sex do/do
inflammation, and repair? Br J Sports Med. 2005;39(5):251–3. not influence post-exercise indexes of muscle damage, inflam-
63. Stupka N, Tiidus PM. Effects of ovariectomy and estrogen on mation, and repair. J Appl Physiol. 2009;106(3):1010–2 (dis-
ischemia–reperfusion injury in hindlimbs of female rats. J Appl cussion 4–15, 21). doi:10.1152/japplphysiol.90848.2008.
Physiol. 2001;91(4):1828–35. 83. Pizza FX, Clark BC, De Meersman RE, et al. Comments on
64. McClung JM, Davis JM, Carson JA. Ovarian hormone status Point:counterpoint: estrogen and sex do/do not influence post-
and skeletal muscle inflammation during recovery from disuse exercise indexes of muscle damage, inflammation, and repair.
in rats. Exp Physiol. 2007;92(1):219–32. J Appl Physiol. 2009;106(3):1016–20. doi:10.1152/japplphysiol.
65. McClung JM, Davis JM, Wilson MA, et al. Estrogen status and 00004.2009.
skeletal muscle recovery from disuse atrophy. J Appl Physiol. 84. Ronkainen PH, Kovanen V, Alen M, et al. Postmenopausal
2006;100(6):2012–23. hormone replacement therapy modifies skeletal muscle compo-
66. McLennan IS. Resident macrophages (ED2- and ED3-positive) sition and function: a study with monozygotic twin pairs. J Appl
do not phagocytose degenerating rat skeletal muscle fibres. Cell Physiol. 2009;107(1):25–33. doi:10.1152/japplphysiol.91518.
Tissue Res. 1993;272(1):193–6. 2008.
67. Massimino ML, Rapizzi E, Cantini M, et al. ED2? macro- 85. Taaffe DR, Sipila S, Cheng S, et al. The effect of hormone
phages increase selectively myoblast proliferation in muscle replacement therapy and/or exercise on skeletal muscle attenu-
cultures. Biochem Biophys Res Commun. 1997;235(3):754–9. ation in postmenopausal women: a yearlong intervention. Clin
68. Enns DL, Tiidus PM. The influence of estrogen on skeletal Physiol Funct Imaging. 2005;25(5):297–304. doi:10.1111/j.
muscle: sex matters. Sports Med. 2010;40(1):41–58. 1475-097X.2005.00628.x.
69. Whiting KP, Restall CJ, Brain PF. Steroid hormone-induced 86. MacNeil LG, Baker SK, Stevic I, et al. 17beta-estradiol atten-
effects on membrane fluidity and their potential roles in non- uates exercise-induced neutrophil infiltration in men. Am J
genomic mechanisms. Life Sci. 2000;67(7):743–57. Physiol Regul Integr Comp Physiol. 2011;300(6):R1443–51.
70. Ghisletti S, Meda C, Maggi A, et al. 17beta-estradiol inhibits doi:10.1152/ajpregu.00689.2009.
inflammatory gene expression by controlling NF-kappaB intra- 87. Dieli-Conwright CM, Spektor TM, Rice JC, et al. Hormone
cellular localization. Mol Cell Biol. 2005;25(8):2957–68. therapy and maximal eccentric exercise alters myostatin-related
71. Pierce AP, de Waal E, McManus LM, et al. Oxidation and gene expression in postmenopausal women. J Strength Condition
structural perturbation of redox-sensitive enzymes in injured Res. 2012;26(5):1374–82. doi:10.1519/JSC.0b013e318251083f.
skeletal muscle. Free Radic Biol Med. 2007;43(12):1584–93. 88. Dieli-Conwright CM, Spektor TM, Rice JC, et al. Influence of
72. Barton ER, Morris L, Musaro A, et al. Muscle-specific expres- hormone replacement therapy on eccentric exercise induced
sion of insulin-like growth factor I counters muscle decline in myogenic gene expression in postmenopausal women. J Appl
mdx mice. J Cell Biol. 2002;157(1):137–48. Physiol. 2009;107(5):1381–8. doi:10.1152/japplphysiol.00590.
73. Tiidus PM, Deller M, Liu XL. Oestrogen influence on myogenic 2009.
satellite cells following downhill running in male rats: a pre- 89. Dieli-Conwright CM, Spektor TM, Rice JC, et al. Hormone
liminary study. Acta Physiol Scand. 2005;184(1):67–72. therapy attenuates exercise-induced skeletal muscle damage in
74. Thomas A, Bunyan K, Tiidus PM. Oestrogen receptor-alpha postmenopausal women. J Appl Physiol. 2009;107(3):853–8.
activation augments post-exercise myoblast proliferation. Acta 90. Dieli-Conwright CM, Spektor TM, Rice JC, et al. HRT and
Physiol (Oxf). 2010;198(1):81–9. mRNA expression of estrogen receptor coregulators following
Sex Hormones and Skeletal Muscle Regeneration

exercise in postmenopausal women. Med Sci Sports Exerc. 108. Craft RM, Clark JL, Hart SP, et al. Sex differences in locomotor
2010;42:422–9. effects of morphine in the rat. Pharmacol Biochem Behav.
91. Dieli-Conwright CM, Spektor TM, Rice JC, et al. Oestradiol and 2006;85(4):850–8. doi:10.1016/j.pbb.2006.11.022.
SERM treatments influence oestrogen receptor coregulator gene 109. Bronstein PM, Wolkoff FD, Levine WJ. Sex-related differences
expression in human skeletal muscle cells. Acta Physiol (Oxf). in rats open-field activity. Behav Biol. 1975;13(1):133–8.
2009;197(3):187–96. doi:10.1111/j.1748-1716.2009.01997.x. 110. Hertrampf T, Degen GH, Kaid AA, et al. Combined effects of
92. Sipila S, Taaffe DR, Cheng S, et al. Effects of hormone physical activity, dietary isoflavones and 17beta-estradiol on
replacement therapy and high-impact physical exercise on movement drive, body weight and bone mineral density in
skeletal muscle in post-menopausal women: a randomized pla- ovariectomized female rats. Planta Med. 2006;72(6):484–7.
cebo-controlled study. Clin Sci (Lond). 2001;101(2):147–57. doi:10.1055/s-2006-931579.
93. Choi SD, Steinberg EM, Lee HH, et al. The timing hypothesis 111. Bowen RS, Turner MJ, Lightfoot JT. Sex hormone effects on
remains a valid explanation of differential cardioprotective physical activity levels: why doesn’t Jane run as much as Dick?
effects of menopausal hormone treatment. Menopause. Sports Med. 2011;41(1):73–86. doi:10.2165/11536860-
2011;18(2):230–6. 000000000-00000.
94. Schierbeck LL, Rejnmark L, Tofteng CL, et al. Effect of hor- 112. Greising SM, Baltgalvis KA, Lowe D, et al. Hormone therapy
mone replacement therapy on cardiovascular events in recently and skeletal muscle strength: a meta-analysis. J Gerontol Ser A
postmenopausal women: randomised trial. BMJ. 2012;345: Biol Sci Med Sci. 2009;64:1071–81.
e6409. doi:10.1136/bmj.e6409. 113. Maltais ML, Desroches J, Dionne IJ. Changes in muscle mass
95. Vingren JL, Kraemer WJ, Ratamess NA, et al. Testosterone and strength after menopause. J Musculoskelet Neuronal Inter-
physiology in resistance exercise and training: the up-stream act. 2009;9(4):186–97.
regulatory elements. Sports Med. 2010;40(12):1037–53. doi:10. 114. Phillips SK, Rook KM, Siddle NC, et al. Muscle weakness in
2165/11536910-000000000-00000. women occurs at an earlier age than in men, but strength is
96. Bhasin S, Storer TW, Berman N, et al. Testosterone replacement preserved by hormone replacement therapy. Clin Sci (Lond).
increases fat-free mass and muscle size in hypogonadal men. 1993;84(1):95–8.
J Clin Endocrinol Metab. 1997;82(2):407–13. 115. Diel P, Friedel A, Geyer H, et al. Characterisation of the phar-
97. Doumit ME, Cook DR, Merkel RA. Testosterone up-regulates macological profile of desoxymethyltestosterone (Madol), a
androgen receptors and decreases differentiation of porcine steroid misused for doping. Toxicol Lett. 2007;169(1):64–71.
myogenic satellite cells in vitro. Endocrinology. 1996;137(4): 116. Gao W, Reiser PJ, Coss CC, et al. Selective androgen receptor
1385–94. modulator treatment improves muscle strength and body com-
98. Bhasin S, Storer TW, Berman N, et al. The effects of supra- position and prevents bone loss in orchidectomized rats. Endo-
physiologic doses of testosterone on muscle size and strength in crinology. 2005;146(11):4887–97. doi:10.1210/en.2005-0572.
normal men. N Engl J Med. 1996;335(1):1–7. 117. Jones A, Hwang DJ, Narayanan R, et al. Effects of a novel
99. Bhasin S, Storer TW, Javanbakht M, et al. Testosterone selective androgen receptor modulator on dexamethasone-
replacement and resistance exercise in HIV-infected men with induced and hypogonadism-induced muscle atrophy. Endocri-
weight loss and low testosterone levels. JAMA. 2000;283(6): nology. 2010;151(8):3706–19. doi:10.1210/en.2010-0150.
763–70. 118. Dalton JT, Barnette KG, Bohl CE, et al. The selective androgen
100. Lee DK. Androgen receptor enhances myogenin expression and receptor modulator GTx-024 (enobosarm) improves lean body
accelerates differentiation. Biochem Biophys Res Commun. mass and physical function in healthy elderly men and post-
2002;294(2):408–13. menopausal women: results of a double-blind, placebo-con-
101. Sinha-Hikim I, Roth SM, Lee M, et al. Testosterone-induced trolled phase II trial. J Cachexia Sarcopenia Muscle.
muscle hypertrophy is associated with an increase in satellite 2011;2(3):153–61. doi:10.1007/s13539-011-0034-6.
cell number in healthy, young men. Am J Physiol Endocrinol 119. Clarke BL, Khosla S. New selective estrogen and androgen
Metab. 2003;285(1):e197–205. doi:10.1152/ajpendo.00370. receptor modulators. Curr Opin Rheumatol. 2009;21(4):374–9.
200200370.2002. doi:10.1097/BOR.0b013e32832ca447.
102. Zhao W, Pan J, Wang X, et al. Expression of the muscle atrophy 120. Runowicz CD, Costantino JP, Wickerham DL, et al. Gyneco-
factor muscle atrophy F-box is suppressed by testosterone. Endo- logic conditions in participants in the NSABP breast cancer
crinology. 2008;149(11):5449–60. doi:10.1210/en.2008-0664. prevention study of tamoxifen and raloxifene (STAR). Am J
103. Liva SM, Voskuhl RR. Testosterone acts directly on CD4? T Obstet Gynecol. 2011;205(6):535 e1–5. doi:10.1016/j.ajog.
lymphocytes to increase IL-10 production. J Immunol. 2011.06.067.
2001;167(4):2060–7. 121. Koot RW, Amelink GJ, Blankenstein MA, et al. Tamoxifen and
104. Cappola AR, Ratcliffe SJ, Bhasin S, et al. Determinants of oestrogen both protect the rat muscle against physiological
serum total and free testosterone levels in women over the age of damage. J Steroid Biochem Mol Biol. 1991;40(4–6):689–95.
65 years. J Clin Endocrinol Metab. 2007;92(2):509–16. doi:10. 122. Grata E, Perrenoud L, Saugy M, et al. SARM-S4 and metabo-
1210/jc.2006-1399. lites detection in sports drug testing: a case report. Forensic Sci
105. Sheffield-Moore M, Paddon-Jones D, Casperson SL, et al. Int. 2011;213(1–3):104–8. doi:10.1016/j.forsciint.2011.07.014.
Androgen therapy induces muscle protein anabolism in older 123. Hayashi SI, Eguchi H, Tanimoto K, et al. The expression and
women. J Clin Endocrinol Metab. 2006;91(10):3844–9. doi:10. function of estrogen receptor alpha and beta in human breast
1210/jc.2006-0588. cancer and its clinical application. Endocr Relat Cancer.
106. Kraemer WJ, Hakkinen K, Newton RU, et al. Effects of heavy- 2003;10(2):193–202.
resistance training on hormonal response patterns in younger vs. 124. Harris HA. The unexpected science of estrogen receptor-beta
older men. J Appl Physiol. 1999;87(3):982–92. selective agonists: a new class of anti-inflammatory agents?
107. Liao YH, Liao KF, Kao CL, et al. Effect of dehydroepiandros- Nucl Recept Signal. 2006;4:e012.
terone administration on recovery from mix-type exercise 125. Velders M, Solzbacher M, Schleipen B, et al. Estradiol and
training-induced muscle damage. Eur J Appl Physiol. genistein antagonize the ovariectomy effects on skeletal muscle
2013;113(1):99–107. doi:10.1007/s00421-012-2409-6. myosin heavy chain expression via ER-beta mediated pathways.
M. Velders, P. Diel

J Steroid Biochem Mol Biol. 2010;120(1):53–9. doi:10.1016/j. 134. Kalbe C, Mau M, Rehfeldt C. Developmental changes and the
jsbmb.2010.03.059. impact of isoflavones on mRNA expression of IGF-I receptor,
126. Weigt C, Hertrampf T, Zoth N, et al. Impact of estradiol, ER EGF receptor and related growth factors in porcine skeletal
subtype specific agonists and genistein on energy homeostasis in muscle cell cultures. Growth Horm IGF Res. 2008;18(5):
a rat model of nutrition induced obesity. Mol Cell Endocrinol. 424–33.
2012;351(2):227–38. doi:10.1016/j.mce.2011.12.013. 135. Jones KL, Harty J, Roeder MJ, et al. In vitro effects of soy
127. Schleipen B, Hertrampf T, Fritzemeier KH, et al. ERbeta-spe- phytoestrogens on rat L6 skeletal muscle cells. J Med Food.
cific agonists and genistein inhibit proliferation and induce 2005;8(3):327–31.
apoptosis in the large and small intestine. Carcinogenesis. 136. Rehfeldt C, Kalbe C, Nurnberg G, et al. Dose-dependent effects
2011;32(11):1675–83. doi:10.1093/carcin/bgr188. of genistein and daidzein on protein metabolism in porcine
128. Hertrampf T, Schleipen B, Velders M, et al. Estrogen receptor myotube cultures. J Agric Food Chem. 2009;57(3):852–7.
subtype-specific effects on markers of bone homeostasis. Mol 137. Serra MC, Beavers KM, Beavers DP, et al. Effects of 28 days of
Cell Endocrinol. 2008;291(1–2):104–8. doi:10.1016/j.mce.2008. dairy or soy ingestion on skeletal markers of inflammation and
03.003. proteolysis in post-menopausal women. Nutr Health.
129. Cvoro A, Tatomer D, Tee MK, et al. Selective estrogen recep- 2012;21(2):117–30. doi:10.1177/0260106012467243.
tor-beta agonists repress transcription of proinflammatory genes. 138. Hubal MJ, Clarkson PM. Counterpoint: estrogen and sex do not
J Immunol. 2008;180(1):630–6. significantly influence post-exercise indexes of muscle damage,
130. Harris HA, Albert LM, Leathurby Y, et al. Evaluation of an inflammation, and repair. J Appl Physiol. 2009;106(3):1012–4
estrogen receptor-beta agonist in animal models of human dis- (discussion 4, 22) doi:10.1152/japplphysiol.90848.2008a.
ease. Endocrinology. 2003;144(10):4241–9. 139. Balagopal P, Olney R, Darmaun D, et al. Oxandrolone enhances
131. Minutolo F, Macchia M, Katzenellenbogen BS, et al. Estrogen skeletal muscle myosin synthesis and alters global gene
receptor beta ligands: recent advances and biomedical applica- expression profile in Duchenne muscular dystrophy. Am J
tions. Med Res Rev. 2011;31:364–442. Physiol Endocrinol Metab. 2006;290(3):e530–9. doi:10.1152/
132. Kuiper GG, Carlsson B, Grandien K, et al. Comparison of the ajpendo.00412.2005.
ligand binding specificity and transcript tissue distribution of 140. Onambele-Pearson GL. HRT affects skeletal muscle contractile
estrogen receptors alpha and beta. Endocrinology. 1997;138(3): characteristics: a definitive answer? J Appl Physiol.
863–70. 2009;107(1):4–5. doi:10.1152/japplphysiol.00448.2009.
133. Pihlajamaa P, Zhang FP, Saarinen L, et al. The phytoestrogen 141. Yager JD, Davidson NE. Estrogen carcinogenesis in breast
genistein is a tissue-specific androgen receptor modulator. cancer. N Engl J Med. 2006;354(3):270–82. doi:10.1056/
Endocrinology. 2011;152(11):4395–405. doi:10.1210/en.2011- NEJMra050776.
0221.

You might also like