Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

Journal of F Jardí et al.

Androgens and physical activity 238:1 R31–R52


Endocrinology

REVIEW

Androgen and estrogen actions on male physical


activity: a story beyond muscle

Ferran Jardí1, Michaël R Laurent2,3, Vanessa Dubois2,†, Nari Kim1, Rougin Khalil1, Brigitte Decallonne1,
Dirk Vanderschueren1 and Frank Claessens2
1Clinical
and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
2MolecularEndocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
3Gerontology and Geriatrics, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium

Correspondence should be addressed to F Jardí: ferran.jardi@kuleuven.be

†(V Dubois is now at INSERM UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France)

Abstract
Physical inactivity is a pandemic that contributes to several chronic diseases and poses a Key Words
significant burden on health care systems worldwide. The search for effective strategies ff physical activity
Journal of Endocrinology

to combat sedentary behavior has led to an intensification of the research efforts to ff exercise
unravel the biological substrate controlling activity. A wide body of preclinical evidence ff muscle
makes a strong case for sex steroids regulating physical activity in both genders, albeit ff brain
the mechanisms implicated remain unclear. The beneficial effects of androgens on ff androgens
muscle as well as on other peripheral functions might play a role in favoring adaptation
to exercise. Alternatively or in addition, sex steroids could act on specific brain circuitries
to boost physical activity. This review critically discusses the evidence supporting a role
for androgens and estrogens stimulating male physical activity, with special emphasis
on the possible role of peripheral and/or central mechanisms. Finally, the potential
Journal of Endocrinology
translation of these findings to humans is briefly discussed. (2018) 238, R31–R52

Introduction
Physical inactivity has emerged as a global health crisis, according to self-reported measures (Hallal et  al. 2012).
being the fourth leading cause of death worldwide Accelerometer data show even lower rates of adherence
according to the World Health Organization (WHO) among adolescents, with only 8% of 12- to 15-year-old
(WHO 2009). Not meeting the minimum physical individuals attaining sufficient physical activity (Troiano
activity recommendations (WHO 2010) is estimated to et al. 2008). Taking into consideration the prevalence and
cause 6% of the burden of disease from coronary heart risks associated with physical inactivity, as well as the
disease, 7% of type 2 diabetes and 10% of both breast and fact that it can be easily and effectively prevented (Heath
colon cancer (Lee et al. 2012). In contrast, 15 min of daily et  al. 2012), it is without surprise that the promotion
moderate-intensity exercise reduces all-cause mortality of physical activity has become a public health priority
by 14% and extends by 3  years the life expectancy (Kohl et al. 2012).
(Wen et  al. 2011). Notwithstanding, one-third of adults The predisposition to engage in physical activity is
worldwide are physically inactive and the prevalence a highly complex trait in humans, probably determined
increases dramatically among adolescents, with four out by the interaction of biological, psychological, social and
of five not reaching the minimum recommended levels environmental factors (Bauman et al. 2012). The role for

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access
Journal of F Jardí et al. Androgens and physical activity 238:1 R32
Endocrinology

a biological substrate influencing the levels of physical running in females is abolished upon gonadectomy and
activity is a relatively recent concept supported by several can be restored following treatment with E2 (Berchtold
lines of evidence, including human studies but mostly et  al. 2001). Based on these and other preclinical data,
preclinical investigations (Bouchard & Rankinen 2006). sex steroids have been suggested as strong candidates
Based on a cohort of 37,000 twin pairs, Stubble et  al. for contributing to the biological substrate influencing
described that the heritability of exercise participation physical activity (Lightfoot 2008). Androgens are well
ranged between 48% and 71% (Stubbe et  al. 2006). known for their potent anabolic actions on muscle and
Nevertheless, the extent to which genetic factors explain we have previously reviewed the cellular targets and
adult exercise behavior is highly heterogenic between molecular pathways implicated (Dubois et  al. 2012).
studies, most likely due to differences in the methodology Therefore, it is conceivable that testosterone increases
used to assess physical activity (Simonen et  al. 2002, activity in males by favoring adaptation to exercise
Eriksson et al. 2006). In addition, candidate gene studies (peripheral mechanisms). In addition or alternatively
have pointed out several hormonal- and neurotransmitter- thereto, testosterone could act on the specific brain neural
related genes associated with physical activity (Stefan et al. circuitries that have been identified to control physical
2002, Salmen et al. 2003, Simonen et al. 2003, Loos et al. activity levels (central mechanisms). Our understanding
2005). However, the two genome-wide association studies of the neural basis underpinning the effects of sex steroids
(GWAS) published so far do not support a prominent role on activity has advanced markedly over the last decade.
for most of these candidate genes (De Moor et  al. 2009, Nevertheless, most of these studies have focused on the
Kim et  al. 2014). This inconsistency might be explained female brain (reviewed in Xu et  al. 2017), whilst the
in part by the limitations of physical activity data based central mechanisms implicated in male activity are less
on questionnaires or diaries, which show poor accuracy documented.
in detecting light-to-moderate activities and are prone to The recent publication of large clinical trials of
error because of individual biases (Strath et al. 2013). The testosterone therapy in elderly men has reignited
latter caveat is overcome by recent efforts to implement the debate on the risks and benefits of testosterone
the use of automated monitors in physical activity replacement (Basaria et  al. 2010, Snyder et  al. 2016,
research. Nonetheless, the strongest body of evidence Storer et  al. 2016). In this context, the positive impact
supporting that physical activity is biologically regulated of testosterone stimulating physical activity and the
still comes from preclinical studies. underlying mechanisms of action represent an additional
Androgens and estrogens are key regulators of consideration for testosterone replacement in elderly,
reproductive organs but also exert multiple effects on mobility-impaired, frail or sarcopenic men. Here, we
non-reproductive tissues, including brain, muscle, adipose review the data supporting a role for androgens and
tissue and bone. Testosterone, the principal circulating estrogens regulating male physical activity and provide
androgen, may stimulate the androgen receptor (AR) an overview of the potential mechanisms implicated.
either directly, or indirectly following 5 alpha-reduction How sex steroids control physical activity in females is
to dihydrotestosterone (DHT), as well as the estrogen reviewed in Lightfoot (2008) and Xu et  al. (2017). We
receptors (ERα and ERβ) after testosterone aromatization will first review some basic concepts regarding physical
into estradiol (E2) (Callewaert et  al. 2010). Even though activity and then discuss several lines of preclinical
research into the biological control of physical activity evidence supporting the notion. In the following sections,
has intensified in recent years, early findings in the 1920s we will review the impact of androgens in the physical
already showed that wheel running in male rats was ability to partake in exercise and examine the specific
abolished after orchiectomy (ORX) (Gans 1927). Later brain circuits that could be underlying the regulation of
studies pinpointed testosterone as the gonadal product activity by testosterone. In the last section, we will briefly
stimulating wheel running, albeit the extent to which discuss what is known regarding the effects of androgens
prior aromatization into E2 was required remained debated and estrogens on physical activity in men.
(see discussion in the ‘Replacement and pharmacological
studies in adult rodents’ section). Physical activity is a
sexually dimorphic trait in rodents (Eikelboom & Mills Categories of physical activity
1988), with females being more active than males and
with fluctuations according to the estrous cycle (Wollnik Physical activity is defined as any bodily movement
& Turek 1988). Similar to what is observed in males, wheel produced by skeletal muscles that results in energy

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access
Journal of F Jardí et al. Androgens and physical activity 238:1 R33
Endocrinology

expenditure (Caspersen et  al. 1985). Although the term animals, thus inducing a substantial psychological stress
‘exercise’ is often interchanged with physical activity, (Sasaki et  al. 2016). In addition, rodents do not always
exercise is indeed a subset of physical activity that is display consistent swimming behaviors and some animals
planned and structured and implies a purpose to achieve will just float, bob and/or dive (American Physiological
health or fitness benefits (Caspersen et  al. 1985). In our Association 2007). Therefore, we focused on voluntary
daily life, we also engage in incidental physical activity wheel running, in which animals have a complete control
when performing home- or work-related activities or on the amount and intensity of exercise.
during transportation (Strath et  al. 2013). Nevertheless,
the latter activities can be performed in an exercise
manner and thus it is not always straightforward to Preclinical evidence supporting a role for
discern between the voluntary (planned) and incidental androgens and estrogens regulating male
(spontaneous) domains of physical activity (Garland physical activity
et  al. 2011). Notably, an increase in energy expenditure
associated with non-exercise activity has been shown Most of our knowledge regarding the role of biological
to prevent fat gain in humans (Levine 1999). Therefore, factors controlling physical activity derives from rodent
physical activity derived from daily tasks may also play a studies and thus the overall significance to human
role in disease prevention and should be included together health remains to be determined. Nevertheless, evidence
with exercise when assessing physical activity. from preclinical studies in rats and mice clearly shows
With regard to preclinical animal studies, wheel the importance of androgens and normal AR and ERα
running in rodents has been extensively used as a signaling in the regulation of male activity.
model of human exercise (Garland et  al. 2011, Novak
et  al. 2012). Similar to the ‘runner’s high’ experienced
Replacement and pharmacological studies in
during endurance exercise in humans, wheel running
adult rodents
is rewarding for rodents and both rats and mice show
preference for environments previously associated with The first observations showing that ORX abrogates wheel
running (Lett et  al. 2001, Fernandes et  al. 2015). When running activity in rats date from almost a century ago
given free access to running wheels, mice reduce the time (Gans 1927). Subsequent studies extended these findings
spent in cage floor activity at the expense of wheel running to mice as well as other rodent species (Daan et al. 1975,
time, the net result being an increase in their total activity Morin & Cummings 1981, Dark & Zucker 1984, Jechura
levels (De Visser et  al. 2005, Silvennoinen et  al. 2014). et al. 2000) and demonstrated the implication of testicular
Spontaneous physical activity (SPA) refers to the non- testosterone production (Daan et  al. 1975, Roy & Wade
exercise component of activity and therefore should not 1975). It is now well established that replacement with
be used as interchangeable for wheel running, although testosterone rescues wheel running activity in a gradual
very often these two parameters correlate reasonably dose-response manner following ORX (Butler et al. 2012).
well. There are several methods to assess SPA in rodents, For instance, the average daily distance run by ORX mice
including infrared photobeams, radiotelemetry, video increases from 1 to almost 4 km following 2 weeks of
tracking systems or force plates (Silvennoinen et al. 2014, treatment with physiological doses of testosterone (Jardí
Teske et al. 2014, Chabert et al. 2016). The main difference et al. 2018). Consequently, the combination of testosterone
between these approaches is their accuracy in detecting and wheel running mediates an adaptational shift to a
ambulation and climbing as well as more subtle stationary more oxidative fiber profile in the skeletal muscle of ORX
movements, such as grooming or feeding. Regardless of the (Allen et  al. 2001, Ibebunjo et  al. 2011). In gonadally
method used, a long (i.e., hours) period of acclimatization intact male rats, administration of supraphysiological
to the activity chamber is required to discharge the doses of testosterone also increases running distances
novelty effects on locomotion (Teske et al. 2014). Studies (McGinnis et al. 2007). Similar to humans, aging in male
failing to meet the former methodological requirement mice is associated with a progressive decline in activity
were excluded from this review. It is important to note (Fig.  1; Hamrick et  al. 2006) and this can be partly
that we focus here on volitional activity, and therefore attenuated by the combination of exogenous testosterone
do not include preclinical studies using forced exercise and low-intensity physical training (Guo et al. 2012). It is
capacity tests. Forced treadmill running or swimming important to note, however, that the concentrations of
require the use of aversive stimuli to motivate the testosterone in male mice appear to remain constant with

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain
Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access
Journal of F Jardí et al. Androgens and physical activity 238:1 R34
Endocrinology

(2.3 km vs. 3.8 km; Jardí et al. 2018) and both testosterone


and DHT were equally effective restoring SPA (Jardí
et  al. 2018). It is noteworthy that DHT metabolites
5α-androstane-3α, 17β-diol (3α-diol) and 5α-androstane-
3β, 17β-diol (3β -diol) may initiate behavioral processes
through AR-independent mechanisms, namely by acting
on gamma-aminobutyric acid (GABA) type A (Frye et  al.
1996) or ERs (Frye et al. 2008a). Nevertheless, we showed
that DHT failed to rescue wheel running in AR knockout
(ARKO) mice, ruling out the implication of AR-independent
Figure 1 mechanisms (Jardí et al. 2018). Additionally, the activity
Changes in SPA with age in male C57BL/6 mice. Mean values are shown
with standard error means as vertical bars. (Data from Hamrick et al.
of ORX+testosterone-treated mice was partially lowered
2006.) by the AR antagonist enzalutamide (Wu et  al. 2016),
further supporting the involvement of AR signaling
aging (Hamrick et al. 2006), in contrast to what is observed in the stimulation of wheel running (Jardí et  al. 2018).
in elderly men (Wu et al. 2008). Thus, it is unlikely that Taken together, these data make a strong case for the
a reduced androgen bioactivity is the main driver of the contribution of AR to the effects of androgens on activity,
decreased SPA in old mice, even when this is responsive at least in male mice, though its importance is somewhat
to testosterone supplementation in the setting of physical less compared to aromatization and ER signaling.
training. In an attempt to investigate the estrogenic component
Many of the actions of testosterone in male behavior of testosterone actions, ORX rats and mice have been
are mediated by estrogen receptors (ERs). For instance, treated with systemic E2 (Roy & Wade 1975, Ogawa et al.
testosterone acts predominantly as a prohormone for E2 in 2003, Bowen et al. 2011, 2012, Blattner & Mahoney 2015,
the programming of male-typical and territorial behavior Royston et al. 2016). It should be noted, however, that the
in mice, although execution of this behavior relies on physiological relevance of this approach is uncertain and
the AR (Wu et al. 2009). It remains debated whether this deserves special consideration. Serum E2 concentrations
dual androgenic and estrogenic action of testosterone in both gonadally intact and ORX+testosterone-, but not
also applies to physical activity. Initially, testosterone was E2-treated mice, are below the limit of detection of sensitive
suggested to stimulate wheel running exclusively through analysis methods using mass spectrometry (McNamara
aromatase-dependent mechanisms (Stern & Murphy 1971, et al. 2010, Nilsson et al. 2015, Laurent et al. 2016b), the
Roy & Wade 1975). Treatment with E2 was more effective ‘gold standard’ for sex steroids measurements. Circulating
than testosterone in stimulating wheel running in ORX levels of E2 are also extremely low in male rats, at the level
rats and the non-aromatizable androgen DHT showed no of a few picograms per milliliter (Quignot et  al. 2012).
effects (Roy & Wade 1975). Also in ORX rats, biologically Therefore, a ‘physiological’ E2 dose is virtually impossible
effective doses of the antiandrogen cyproterone acetate to define in male rodents, and most ‘replacement’ doses
failed to inhibit the stimulatory actions of testosterone are almost by definition supraphysiological. Remarkably,
on wheel running (Stern & Murphy 1971). In accordance, aromatase is highly expressed in several brain regions
treatment with the 5α-reductase inhibitor dutasteride did of male rodents, including the bed nucleus of the stria
not alter the levels of wheel running in male Zucker obese terminalis, the olfactory tubercle, the medial amygdala
rats (Sato et al. 2013). In the latter study, however, the lack (MeA) and the hypothalamus, suggesting a role for the
of effects could be potentially explained by the already autocrine and paracrine actions of estrogens (Zhao
low levels of wheel running of these animals (Stern & et  al. 2007, Stanić et  al. 2014). Overall, we cannot draw
Johnson 1977) and/or by a compensatory effect related conclusions about the physiological role of E2 on physical
to the secondary increases in testosterone concentrations activity levels based on studies treating male rodents with
(Amory et  al. 2007). Later studies challenged the notion systemic E2, even when pharmacological doses of E2
of aromatase being indispensable for testosterone effects restore wheel running following ORX (Roy & Wade 1975,
on wheel running. In male mice, treatment with DHT Ogawa et  al. 2003, Bowen et  al. 2011, 2012, Blattner &
partly restored the drop in wheel running following Mahoney 2015, Royston et al. 2016). Although treatment
ORX (Karatsoreos et  al. 2007, Iwahana et  al. 2008), with the aromatase inhibitors letrozole and exemestane
although to a lesser extent compared with testosterone did not affect wheel running either in gonadally intact

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access
Journal of F Jardí et al. Androgens and physical activity 238:1 R35
Endocrinology

or ORX+testosterone-treated mice (Bowen et  al. 2011), The reduction of SPA following ORX, however, was only
this study needs further technical validations. The partially rescued by MK-4541 (Chisamore et al. 2016). In
ultimate proof of the role of ERs on male physical activity a similar pattern, treatment with SARM-2f completely
comes from studies in knockout mice lacking ERα or restored muscle mass in ORX mice but was less effective
the aromatase enzyme (see discussion in the ‘Global KO increasing their wheel running activity (Morimoto
mouse models’ section). et  al. 2018). In a preclinical mouse model of muscular
dystrophy, an enobosarm analog extended the survival of
Androgenic-anabolic steroids (AAS) and selective mice but failed to increase their low SPA (Ponnusamy et al.
androgen receptor modulators (SARMS) 2017). Nevertheless, the severity of the pathology of these
AAS are synthetic derivatives of testosterone that were mice may have impeded an increase in their SPA levels.
developed in an attempt to maximize the anabolic The effects of SARMs on physical activity are worthy of
activity of testosterone. The prolonged abuse of AAS further study and could represent an additional benefit in
in men is related to several psychiatric adverse effects, the treatment of muscle wasting conditions.
including irritability, mood swings, mania (‘steroid
rage’ at supraphysiological doses) and depression
Perinatal studies
(Pope & Katz 1994, Bjørnebekk et  al. 2017). Although
scarce, a few preclinical studies have explored whether So far, we discussed how androgens stimulate physical
doses of AAS that model human abuse patterns affect activity in adult rodents. It is noteworthy that behavioral
physical activity levels. In contrast to testosterone, both responses to sex steroids during adulthood are influenced
nandrolone (McGinnis et al. 2007, Tanehkar et al. 2013) by sex steroids acting on the nervous system during early
and stanozolol (McGinnis et  al. 2007) reduced wheel critical periods. In their seminal work in 1959, Phoenix
running activity in gonadally intact male rats. Similarly, et  al. were the first to dichotomize hormonal effects in
the combination of nandrolone and methandrostenolone two groups: organizational vs. activational (Phoenix et al.
decreased voluntary wheel running in mice (Onakomaiya 1959). The organizational-activational theory states that
et al. 2014). Paradoxically, despite the detrimental effects hormones sculpt neural tissues during development so
of nandrolone on wheel running in male rats (McGinnis that they respond differentially to hormonal activation
et al. 2007, Tanehkar et al. 2013), the combination of both in adulthood (Phoenix et  al. 1959). The study of the
interventions improved their running endurance (Vanzyl organizational effects of sex steroids has mainly focused
et  al. 1995). As for the mechanisms of action, AAS and on the programming of mating behavior (Arnold 2009).
endogenous testosterone differ in both the affinity to Although less evidence is available for non-sexual
bind AR and the ability to be aromatized (Bergink et  al. behavior, the results from several preclinical studies
1985, Fragkaki et al. 2009). Possibly, AAS regimes reduce suggest that the perinatal sex steroid milieu might also
wheel running by interfering with the endogenous determine physical activity in adulthood.
testosterone production and hence E2 (Barone et  al. The perinatal surges of testosterone in male rodents
2017). Alternatively, AAS might act directly on several during the late embryogenic period and the first hours
neurotransmitter systems in the brain that affect physical of life are critical for the male brain to become capable
activity (Mhillaj et al. 2015). of producing male sexual behavior (masculinization)
The need to improve both the benefit/risk profile and whilst losing its ability to respond in a female-like pattern
pharmacokinetics of androgens spurred the development (defeminization; Lenz & McCarthy 2010 for review on this
of selective androgen receptor modulators (SARMs) topic). Apart from the perinatal period, a growing body of
(Mohler et  al. 2009). SARMs received initially a great evidence shows that during puberty, testosterone is also
deal of clinical attention for their potential use in the required for the full masculinization and defeminization
treatment of muscle wasting conditions. To date, yet, none of male brain (Schulz et al. 2004, De Lorme & Sisk 2016,
of these compounds has shown unequivocal efficacy in Sano et  al. 2016). Paradoxically, the organizational
clinical trials (Almeida et al. 2017). Preclinical data show effects of testosterone on the developing male brain are
that SARMs display a better myotrophic index compared actually driven by ERs, suggesting the implication of
to their steroidal analogues. In ORX mice, treatment brain aromatase activity (Ogawa et  al. 2000, Wu et  al.
with the SARM MK-4541 led to an increase in muscle 2009, Juntti et al. 2010). In females, prepubertal estrogen
mass and function comparable to DHT but without the levels are very low, and the traditional dogma states
sparing effects on seminal vesicles (Chisamore et al. 2016). that sexual behavior is merely the result of the absence

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain
Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access
Journal of F Jardí et al. Androgens and physical activity 238:1 R36
Endocrinology

of perinatal testosterone organizational actions. Recent treatment (Royston et al. 2016). From these observations,
data challenge the latter hypothesis demonstrating that it is reasonable to assume that the neonatal testosterone
E2 actions outside the perinatal period are required for surge programs male physical activity and that this
the normal development of female sexual behavior (Brock might involve aromatase-dependent processes (Fig.  2).
et al. 2011). The neural substrate underlying the persistent In support of this premise, female rats treated neonatally
and irreversible effects of sex steroid actions on the with androgens develop a resistance to E2 stimulation of
developing brain remains mostly unknown. Although not wheel running in adulthood, which can be prevented by
an exhaustive list, some suggested mechanisms include treating pups with antisense oligodeoxynucleotides to
epigenetic modulation (Nugent et al. 2015), regulation of ER mRNA (Blizard 1983, McCarthy et  al. 1993, Royston
cell death and proliferation (Wu et al. 2009) and changes et al. 2016). Whether the neural mechanisms underlying
in synaptic patterning (Schwarz et al. 2008). the organizational effects of testosterone on activity are
Activity is a sexually dimorphic behavior in rodents, similar to those described for mating behavior remains
with females showing higher levels of wheel running and to be investigated. Similarly, the organizational effects
SPA compared to males (Eikelboom & Mills 1988). Gender of testosterone outside the perinatal window on physical
differences in activity remain after gonadectomy and activity (i.e. puberty) await confirmation.
thus cannot be explained by hormonal dimorphism in
adulthood (Gentry & Wade 1976, Blizard 1983, Broida &
Global KO mouse models
Svare 1984, Bowen et al. 2012, Kuljis et al. 2013). Differences
in sex chromosomes as such are also insufficient to explain Studies in transgenic mice lacking either steroid hormone
the dimorphism in activity, as evidenced by studies using receptors or the aromatase enzyme provide further insights
the mouse model of 4 core genotypes, in which sex into the role of sex steroids regulating physical activity.
chromosome complement is independent of gonadal However, the interpretation of the results obtained from
phenotype (De Vries et  al. 2002, Kuljis et  al. 2013). In a these mouse models is hampered by alterations of the
similar pattern to XX females, ovariectomy of mice with a hypothalamic–pituitary–gonadal (HPG) axis. In addition,
deletion of the testis-determining gene Sry results in running it is reasonable to expect changes in the sex-steroid-
longer distances than ORX males (Kuljis et al. 2013). dependent brain organization (see previous section)
Accumulating literature suggests a role for sex steroid caused by the gene deletion during development.
actions during the perinatal period determining gender
differences in activity. ORX at postnatal day 0 (P0) Androgen receptor knockout (ARKO) mice
results in higher levels of SPA than that at P5, 10 or 25 Several ARKO mouse models targeting either exon 1, 2 or 3
in male mice (Broida & Svare 1984). Also in male mice, have been generated using the Cre-LoxP system (Fan et al.
estrogen deficiency during early life leads to a higher 2005, Ophoff et  al. 2009a, Rana et  al. 2011). Our group
wheel running response in adulthood following E2 reported an almost complete abrogation of wheel running

Figure 2
Schematic overview of the overall proposed
mechanisms of action of testosterone on male
activity (see text). A, androgens; CNS, central
nervous system; E2, estradiol; PNS, peripheral
nervous system; +, stimulatory effect; ++,
dominant stimulatory effect.

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access
Journal of F Jardí et al. Androgens and physical activity 238:1 R37
Endocrinology

as well as a reduced SPA in exon 2-deleted male ARKO mice show increased levels of serum testosterone (Callewaert
(Ophoff et al. 2009a). A similar low activity phenotype was et  al. 2009), are less active than their wild-type (WT)
observed in mice with a targeted disruption of AR exon 1 littermates when given access to running wheels (Blattner
(Fan et al. 2005) and in a DNA-binding domain ARKO mouse & Mahoney 2012), and develop elevated adiposity in late-
model that only retains non-genomic actions (Rana et  al. life (Heine et al. 2000). As shown first in the seminal work of
2011). The fact that global ARKO mice are less physically Ogawa et al. and later confirmed by others, the stimulatory
active might predispose them to develop late-onset actions of E2 treatment on wheel running activity are
adiposity (Fan et  al. 2005, Rana et  al. 2011). Global ARKO abrogated in ERαKO mice (Ogawa et al. 2003, Dworatzek
mice appear to suffer from an impaired gonadal testosterone et al. 2014, Blattner & Mahoney 2015). This is not the case
production (Callewaert et al. 2009) and might therefore not for ERβKO mice in which the response to E2 is unaffected
be useful to discern between purely AR-mediated effects and (Ogawa et  al. 2003). Therefore, the estrogenic regulation
those secondary to E2 deficiency. Indeed, treatment with of wheel running in males is primarily mediated by ERα,
testosterone completely restored wheel running in male though this does not exclude the possibility that ERβ plays
ARKO mice, an effect associated with a normalization of a role in modulating the circadian-rhythm patterns of
their E2 concentrations in brain (Jardí et al. 2018). Thus, it activity, as shown for females (Royston et al. 2014).
is reasonable to assume that the low activity phenotype of
ARKO mice is derived in large part from a lack of testosterone ‘Non-classical’ ER knock-in (NERKI) mice
substrate for aromatization into E2. Transgenic mice with a knock-in mutation in the ERα
selectively abolishing estrogen response element (ERE)
Aromatase knockout mice binding were originally generated to distinguish between
Mice in which E2 biosynthesis is disrupted by deletion classical and non-classical ERα actions in vivo (Jakacka
of the Cyp19a1 gene represent a useful tool to study the et al. 2002). Male ‘non-classical’ ER knock-in mice (NERKI)
implication of E2 in the regulation of male physiological mice show a reduced wheel running activity that does not
processes (Cooke et  al. 2017). Most studies in male increase following challenge with E2 (Blattner & Mahoney
aromatase KO mice agree that both wheel running (Watai 2012, 2015). Based on these observations, it is tempting to
et  al. 2007, Brockman et  al. 2011) and SPA (Hill et  al. speculate that ERα actions on wheel running activity are
2007) are diminished in the absence of E2. Nevertheless, dependent on ERα binding to ERE elements.
one study reported increased levels of wheel running in
male aromatase KO mice, as part of a broader spectrum of G-protein-coupled receptor 30 knockout
compulsive behaviors (Hill et al. 2007). It should be noted (GPR30KO) mice
that the authors analyzed pre-plateau phases of activity, The G-protein-coupled receptor 30 (GPR30) has been
which could have led to a mistaken estimation of the proposed as a membrane ER (Langer et  al. 2010). There
‘true’ levels of wheel running. Be that as it may, aromatase are important phenotypic differences between the four
KO mice also show an impaired negative feedback of the independent GPR30KO mouse models available, most
HPG axis, with higher levels of serum testosterone and likely reflecting that other factors apart from deletion of
gonadotropins (Fisher et  al. 1998, Jones et  al. 2000, Öz GPR30 are contributing to the observed effects (Langer
et  al. 2000). The fact that aromatase KO mice display a et  al. 2010). In two different GPR30KO mouse models,
low activity phenotype even in the presence of excess males exhibited normal SPA levels (Sharma et  al. 2013,
testosterone indicates a crucial role for aromatization in Kastenberger & Schwarzer 2014), but another study reported
the testosterone-induced stimulation of male physical that GPR30KO male mice displayed a mild reduction
activity. Supporting this notion, male aromatase KO mice in wheel running as well as an impaired left-ventricular
do not show an ORX-induced reduction of wheel running cardiac function (Delbeck et al. 2011). It therefore remains
(Brockman et al. 2011). uncertain whether GPR30 is physiologically relevant for
physical activity, and if so, if it contributes to E2-induced
Estrogen receptor α (ERα-) and β knockout responses and/or initiates independent mechanisms.
(ERβKO) mice
The generation of ERα and ERβKO mice has advanced Sex-hormone-binding globulin transgenic
our understanding of the mechanisms implicated in the (SHBG-tg) mice
estrogenic regulation of male physiology (Cooke et  al. Sex-hormone-binding globulin (SHBG) is a high-affinity
2017). Similar to male aromatase KO mice, ERαKO mice binding protein for testosterone, DHT and E2 that is found

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain
Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access
Journal of F Jardí et al. Androgens and physical activity 238:1 R38
Endocrinology

circulating in humans but is almost completely lacking in et al. 2012) and, in turn, the capacity to exercise. In other
rodent serum. We recently showed in a humanized mouse words, testosterone might influence both the motivation
model expressing human SHBG that testosterone needs and/or the ability to partake in activity, referred here as
to be biologically available (i.e. not bound to SHBG) to central and peripheral actions, respectively (Fig. 2). Note
produce its stimulatory effects on wheel running (Jänne that the term ‘peripheral’ as applied to this classification
et  al. 1998, Jardí et  al. 2018). In SHBG-tg mice, the does not preclude the implication of the central nervous
majority of testosterone is bound by SHBG and cannot system (CNS) in the observed effects.
enter target tissues by the canonical way (Laurent et  al.
2016a,b). Male SHBG-tg mice display features of a mild
Peripheral actions
androgen deficiency phenotype, including a 50% decrease
in wheel running activity (Jardí et al. 2018). As the decline In the next section, we will examine whether androgens
of androgen bioactivity in elderly men is also only mild provide greater tolerance to exercise by improving muscle
(Wu et  al. 2008), SHBG-tg mice may be a more suitable strength, motor skills, metabolism and/or other peripheral
animal model to study late-onset hypogonadism than parameters in male rodents.
ORX.
The interpretation of studies using constitutive KO Muscle mass and strength
mice for steroid hormone receptors or the aromatase Both the mass and the ex vivo contractility of hindlimb
enzyme is subject to several limitations, including muscles show a mild reduction following ORX or AR
alterations of the HPG axis, developmental compensatory deletion in male mice (Axell et  al. 2006, MacLean et  al.
mechanisms and the impossibility to distinguish tissue- 2008, De Naeyer et  al. 2014, Dubois et  al. 2014). The
specific contributions. Nonetheless, findings in both differences in strength disappear when normalized for
aromatase and ERKO mice indicate that testosterone muscle weight, suggesting that the intrinsic contractile
actions on physical activity require normal aromatase and properties of muscle remain unaltered (Axell et al. 2006,
ERα functions. In contrast, we cannot draw conclusions MacLean et al. 2008, Hourdé et al. 2009, De Naeyer et al.
about the implication of AR from studies in global ARKO 2014). However, in five different myoblast or satellite-
mice because of their severe hypogonadism. However, cell-specific ARKO models, the mass and contractility
replacement and pharmacological studies made clear of hindlimb muscles were not, or marginally, affected
that the contribution of AR to testosterone effects cannot (Ophoff et al. 2009b, Chambon et al. 2010, Dubois et al.
be dismissed as minimal or trivial (see discussion in 2014, Ferry et  al. 2014, Rana et  al. 2016), whilst all five
the ‘Replacement and pharmacological studies in adult models showed a partial decrease in the weight of the
rodents’ section). androgen-sensitive perineal muscles. Nevertheless, the
expression of AR in the muscle cell lineage was required
for the full gain in muscle mass induced by mechanical
Mechanisms implicated in the effects of loading (Ferry et al. 2014), an effect that could be related
androgens and estrogens on male to the modulation of a reserve pool of adult muscle stem
physical activity cells by androgens (Kim et  al. 2016). An overview of
the main features of the different muscle-specific ARKO
Despite the large amount of preclinical data showing models is presented in Table 1. Overall, the former studies
that androgens and estrogens control physical activity suggest a role for non-myocytic AR mediating the modest
in males, the mechanisms involved are not well actions of androgens on murine muscle. In this regard,
understood. Due to their pleiotropic nature, sex steroids a recent study points toward neuronal AR contributing
could stimulate physical activity by acting on multiple to maintain hindlimb muscle mass but not contractility
organs or systems. Of particular interest, the use of in male mice (Davey et  al. 2017). The cellular targets
genetic techniques to manipulate neuronal pools in a underlying the anabolic actions of androgens on muscle
selective manner has allowed the identification of several have been reviewed by Dubois et al. (2012).
sex-steroid-responsive neural circuitries implicated in The ex vivo contractility studies referred to in the
the drive to engage in physical activity (Musatov et  al. earlier sections do not support a role for an intrinsic
2007, Xu et  al. 2011, 2015, Correa et  al. 2015). Besides muscle dysfunction impairing the physical ability of ORX
these central mechanisms, androgens could also increase and ARKO mice. Yet, these findings do not exclude the
activity levels by improving muscle function (Dubois possibility that androgens regulate the neuromuscular

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access
Journal of F Jardí et al. Androgens and physical activity 238:1 R39
Endocrinology

function in vivo. Exercise training potentiates the

lox hemizygous mice show a phenotype of hyperandrogenization; bAR expression also reduced in brain and fat; cas assessed by SV weight; dtreadmill running; edetermined by DXA; frotarod test;
SPA

ND
ND
ND
ND
ND
ND

ND
ND

ND
(↑)


stimulatory effects of androgens on muscle mass and

↑, increased; (↑), SPA restored to control values following normalization of testosterone levels; ((↑)), not statistically significant; ↓, decreased; =, unchanged; (=), minor impact on fatigue but no
strength (Guo et al. 2012, Cozzoli et al. 2013). Indeed, the

changes in maximal muscle tension; ND, not determined; neu, neurons; sat, satellite cells; skm, skeletal muscle myofibers; SPA, spontaneous physical activity; SV, seminal vesicles; testosterone,
running
Wheel

ND combination of both interventions benefits endurance


ND
ND

ND
ND
ND
=
=

=

capacity in rodents (Vanzyl et  al. 1995, Georgieva &
Boyadjiev 2004, Cozzoli et al. 2013), presumably through
increasing muscle function. However, ORX per se
performance
Motor

diminishes grip strength by only about 5–15% in some


ND
ND
ND
ND

ND

ND
ND
ND
=d
=d

=f but not all studies (Borst et al. 2007, Windahl et al. 2011,
White et al. 2013, Chisamore et al. 2016). The discrepancies
between studies might be due to the lack of sensitivity of
Contractile
function

the conventional forelimb grip strength test (Takeshita


ND
ND

ND

ND
ND
ND

ND
(=)
(=)

=

et al. 2017). Additionally, the motivation to hold onto the


grid may vary between mice, causing inconsistencies in
the results. Be that as it may, a modest reduction (~10%) in
strength

grip strength was also found in two muscle-specific ARKO


Grip

ND
ND

ND

ND
ND
ND
ND
=

mouse models (Chambon et al. 2010, Dubois et al. 2014)


(Table  1). However, this did not impede muscle-specific
ARKO mice from displaying similar performances to their
Muscle
mass

ND
ND

WT littermates in both the voluntary wheel running and


=e
=
=
=

=

the forced treadmill tests (Table 1). Overall, we conclude


that it is unlikely that the mild loss of muscle function
Serum testosterone/SV weight

following androgen deficiency in male mice is the main


driver of their low activity phenotype.

Motor skills
((↑))
ND
ND
=c
=c

↑c

↑c
=

Preclinical data show that testosterone deficiency exerts


detrimental effects on male CNS health (Khasnavis et al.
Table 1  Overview of muscle- and neuronal-specific ARKO mouse models.

2013, Jayaraman et al. 2014, Atallah et al. 2017). In mice,


prepubertal ORX led to a poor performance in the rotarod
test, which the authors associated with a loss of midbrain
dopaminergic neurons (Khasnavis et al. 2013). Conversely,
Dubois et al. (2014, 2015),

also in the rotarod test, male testicular-feminized (Tfm)


Chambon et al. (2010)
Chambon et al. (2010)
Ophoff et al. (2009b)

mice expressing a non-functional AR performed better


Raskin et al. (2009)

Davey et al. (2017)


Jardí et al. (2017b)
Jardí et al. (2018)

Juntti et al. (2010)


Rana et al. (2016)
Rana et al. (2016)

than their WT littermates (Rizk et  al. 2005). Although


Yu et al. (2013)

this might seem unexpected at first, the increased rotarod


performance of Tfm mice could be due to their lower
body weight, since these two variables share an inverse
correlation (Mao et al. 2015). Confirming the latter results,
Ref

expression retained in the hypothalamus.

male ARKO mice showed normal motor skills despite


their loss in striatal dopamine concentrations, a fact that
ARNeu;Thy1CreER(g)

might be explained by compensatory adaptations during


ARNeu;SynapsinICre
ARskm;HSACre(a,b)

ARNeu+glia;NesCre
ARNeu+glia;NesCre

ARNeu;CamKIIαCre
ARskm;MCKCre(a)

ARsat+skm;MyoD
ARskm;HSACreER
Mouse strain

ARskm;MCKCre

ontogeny (Jardí et  al. 2018). In contrast to prepubertal


ARskm;HSACre

castration, ORX adult rodents showed an altered


performance in neither the rotarod test nor a nigrostriatal
pathology (Kritzer et al. 2001, Frye et al. 2008b, Khasnavis
et al. 2013). In the beam-walking test, which detects more
testosterone.
Myofibers

subtle motor coordination deficits, one study showed that


Cell type

Neurons

ORX mice performed worse than sham-operated controls


gAR
aAR

(McDermott et  al. 1994). However, the performance in

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain
Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access
Journal of F Jardí et al. Androgens and physical activity 238:1 R40
Endocrinology

the latter test is in part influenced by motivational factors maintaining the capacity of male rodents for endurance
(McDermott et al. 1994, Curzon et al. 2009, Deacon 2013). activities. In contrast to what is observed in the voluntary
Regarding peripheral nerve health, both ORX and wheel running test, both ARKO mice and ORX rats
treatment with the antiandrogen flutamide reduced perform as well as control animals when forced to run
the synthesis of myelin proteins in the sciatic nerve of in a treadmill (Ophoff et al. 2009a, Petroianu et al. 2010).
male rodents (Magnaghi et  al. 1999, 2004, Jayaraman Therefore, we conclude that androgens must act through
et al. 2014). In male mice, the decrease in myelin sheath other mechanisms beyond muscle to stimulate physical
was associated with macrophage infiltration (Jayaraman activity. Nevertheless, it is still possible that the increase
et al. 2014). Also in male mice, however, nerve function in activity over time potentiates the peripheral actions of
as assessed by sciatic nerve conductance studies was not androgens so that they acquire a greater importance in
altered following either ORX or ARKO (Jardí et al. 2018). sustaining activity (Fig. 2).
Taken together, we conclude that the adverse actions
of androgen deficiency on male neural health do not seem
Central actions
to translate into a deterioration of motor performance
that would limit physical activity, at least in adult rodents. In the next section, we will review the available findings
This does not preclude a role for androgens improving supporting the notion that testosterone is mainly acting
motor functional recovery in animal models of neural centrally to boost physical activity in male rodents.
disease (Uchida et al. 2009, Yoo & Ko 2012, Ponnusamy
et al. 2017). Central-nervous-system-specific KOs
In the mouse brain, AR and ERα display a moderate-to-
Other peripheral actions high regionalized expression profile, both showing a high
Skeletal muscle metabolism is critical to maintain fuel expression in the hypothalamus and brainstem, whilst AR
homeostasis during exercise. In both male rats and mice, levels are also abundant in the hippocampus and cerebral
androgen deficiency decreases muscle glycogen stores cortex (Gofflot et al. 2007, Lein et al. 2007, Mahfouz et al.
(Ramamani et  al. 1999, Dubois et  al. 2016), a suggested 2016). Four different whole brain ARKO mouse models
risk factor for fatigue development (Xirouchaki et  al. have been generated so far, allowing to explore the impact
2016). Conversely, testosterone promotes glucose uptake of AR signaling in the nervous tissue without interfering
in skeletal muscle, although it is not clear to what extent with its peripheral functions (Raskin et  al. 2009, Juntti
these effects are dependent on local AR actions (Sato et al. et al. 2010, Yu et al. 2013, Davey et al. 2017). An overview
2008, Ibebunjo et al. 2011, Dubois et al. 2016, Kelly et al. of the main features of the different neuronal ARKO
2016). In male mice, deletion of myogenic AR did not models is presented in Table  1. All four models show
affect either muscle glycogen stores (Ophoff et al. 2009b) an intact peripheral masculinization but unfortunately
or glucose tolerance (Dubois et al. 2016), in contrast to that have an impaired negative feedback of the HPG axis in
observed in both liver- (Lin et al. 2008) and adipose-tissue- males, resulting in higher serum levels of testosterone
specific ARKO mice (McInnes et al. 2012). The mechanisms and increased seminal vesicle weights (Raskin et al. 2009,
by which testosterone acts on metabolic pathways have Juntti et  al. 2010, Yu et  al. 2013, Davey et  al. 2017). It is
been reviewed elsewhere (Kelly & Jones 2013). Besides nevertheless clear that the neuronal AR is required for the
metabolism, long-term ORX might affect exercise full expression of both male-typical sexual and aggressive
capacity in rodents by attenuating cardiac contractility, behaviors (Raskin et al. 2009, Juntti et al. 2010, Marie-Luce
as suggested by both in vivo echocardiographic and ex et al. 2013). Regarding physical activity, one study reported
vivo studies (Sebag et al. 2011, Eleawa et al. 2013). These an increase in SPA in Nestin-Cre neuronal ARKO mice,
alterations in cardiac contractile function could arise from although levels were restored to control values following
testosterone modulating Ca2+ handling in ventricular normalization of testosterone levels, pointing to an
myocytes, as recently reviewed (Ayaz & Howlett 2015). implication of the altered HPG axis in the observed effects
Additionally, androgens might influence adaptation to (Raskin et al. 2009). Contrarily, both SPA and wheel running
exercise by affecting hemoglobin and hematocrit levels were reduced by 60% in CamKII-Cre neuronal ARKO
(Bhasin et  al. 2012), bone (Vanderschueren et  al. 2014) mice, which retains only non-DNA-binding-dependent
and/or lung function (Townsend et  al. 2012). However, AR actions (Davey et  al. 2017). The latter results support
overall, the actions of androgens on these parameters or the interpretation that AR exerts a predominant role in
on muscle do not appear to play an indispensable role in the regulation of male physical activity by testosterone.

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access
Journal of F Jardí et al. Androgens and physical activity 238:1 R41
Endocrinology

Nonetheless, the above-mentioned studies did not include to estrogens in females, regulate male energy expenditure
littermates carrying the Cre transgene as controls. This is by acting on AR and/or ER-positive hypothalamic
particularly important since Cre-driven transgenes might neurons. Hypothalamic testosterone implants restored
exert independent effects on behavioral outcomes (Harno wheel running activity in ORX mice (Model et al. 2015).
et al. 2013, Chen et al. 2016). There is also the possibility However, since testosterone can diffuse to adjacent brain
that brain AR deletion at early stages may have indirectly areas, these might also be implicated in the observed
interfered with the organizational actions of testosterone effects. According to unpublished data of Ogawa et  al.
by altering the HPG axis. To circumvent this caveat, we (Sano et  al. 2013), site-specific ERα knockdown in the
recently generated a tamoxifen-inducible neuronal ARKO VMH completely abolished estrogen-induced facilitation
mouse model by using the Thy1-CreER, which drives gene of wheel running in male mice, similar to that described
deletion in extrahypothalamic regions of the brain (Jardí in females (Fig. 3). Notwithstanding the aforementioned
et  al. 2017a). In contrast to CamKII-Cre neuronal ARKO limitation of treating male mice with estrogens, these
mice, deletion of AR in the CNS of pubertal male Thy1- results suggest that the VMH forms part of the neural
CreER neuronal ARKO mice did not affect wheel running circuitries mediating testosterone-induced wheel running
(Jardí et al. 2017b). The differences between studies could in male mice and highlights the implication of local
be attributed to the residual expression of hypothalamic aromatization in this brain region (Roselli et  al. 1985).
AR in our mice (Jardí et al. 2017a). All in all, the study of In contrast to females, however, the regulation of male
neuronal ARKO mice has not provided clear answers so far physical activity by ERα expression in the VMH may
regarding the role of brain androgenic signaling in male involve a distinct module than the ventrolateral region
physical activity. To the best of our knowledge, only one since stimulation of this neuronal cluster did not increase
study reported a whole-brain ERαKO in male mice, but the SPA in male mice (Correa et al. 2015).
authors did not assess physical activity in these animals
(Xu et al. 2011). Medial amygdala
The neuronal basis regulating physical activity
Hypothalamic circuitries likely implies other neuronal circuitries beyond the
The hypothalamus is a key brain region that integrates hypothalamic energy balance systems, such as brain
nutritional, hormonal and neural information to regions involved in emotional and motivational
orchestrate adaptive physiological responses aimed to processing (Fig.  3). MeA neurons projecting to the
maintain the whole-body energy balance (Lenard & hypothalamus play a central role in generating emotional
Berthoud 2008, Schneeberger et  al. 2014). Technical responses to chemosensory signals (Keshavarzi et  al.
advances in viral-vector-mediated gene manipulation 2014, Takahashi 2014). Compared to other amygdalar
as well as in opto- and chemogenetics have allowed the nuclei, the expression of sex steroid receptors is enriched
identification of hypothalamic circuitries implicated in the MeA (Gofflot et  al. 2007). In addition, a high
in the control of energy homeostasis by sex steroids. proportion of MeA neurons are aromatase positive, and
In particular, hypothalamic ERα signaling in female therefore this brain region might be exposed to relatively
rodents has been shown to exert a holistic control of high levels of E2 (Wu et al. 2009, Unger et al. 2015). In
their overall energy balance, influencing food intake as male mice, ERα signaling in the MeA is required for the
well as energy expenditure (Xu et al. 2017 for review on pubertal organizational actions of testosterone on social
this topic). Silencing of ERα in the ventromedial nucleus and sexual behaviors (Sano et al. 2016). In addition, Xu
of the hypothalamus (VMH) by small hairpin RNA et  al. provided compelling evidence that ERα signaling
knockdown led to obesity in both female rats and mice, in the MeA stimulated SPA in males and secondarily
an effect associated with a decline in physical activity prevented body weight gain in the setting of high-fat diet
and thermogenesis (Musatov et  al. 2007). Remarkably, (Xu et al. 2015). Particularly, deletion of ERα in the MeA
the effects of estrogens on these two parameters of of adult male mice resulted in a low activity phenotype,
energy expenditure seem to be driven by independent whilst the other components of energy expenditure,
ERα modules of the VMH (Xu et  al. 2011, Correa et  al. namely thermogenesis and resting metabolic rate,
2015). In male mice, global deletion of either AR or ERα remained unaltered (Xu et  al. 2015). Together with
results in a decreased energy expenditure that might the VMH (unpublished data of Ogawa et  al.), the MeA
predispose them to increased adiposity (Heine et al. 2000, is the only brain region identified so far that could be
Fan et al. 2005). It is not clear whether androgens, similar mediating the stimulatory effects of testosterone on

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain
Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access
Journal of F Jardí et al. Androgens and physical activity 238:1 R42
Endocrinology

male physical activity (Fig. 3). In addition, these findings receptors but also indirectly through the afferent neurons
also imply a role for brain aromatization in testosterone projecting to them (Kritzer & Creutz 2008, Aubele &
regulation of activity, similar to that described for male- Kritzer 2012, Purves-Tyson et  al. 2012, Locklear et  al.
typical aggressive behavior (Unger et al. 2015). 2017). However, it is not clear if the main contribution of
androgens to the overall dopaminergic tone is at the level
Rewarding and motivational pathways of dopamine synthesis, transport, release, metabolization
Besides the metabolic-regulatory circuitries of the and/or signaling (Aubele & Kritzer 2011, Khasnavis
hypothalamus and the MeA, brain regions controlling et  al. 2013, Purves-Tyson et  al. 2014, Wang et  al. 2016).
rewarding and motivational functions might also be Testosterone restoration of basal extracellular dopamine
involved in the regulation of physical activity (Garland levels in the mesocortical dopaminergic pathway implies
et  al. 2011) (Fig.  3). Endurance running generates a AR-dependent mechanisms, though this might not apply
rewarding aftereffect in both humans and rodents to the rest of dopaminergic systems (Kritzer 2003, Aubele
(Boecker et al. 2008, Fernandes et al. 2015). The traditional & Kritzer 2011, Locklear et  al. 2017). Amphetamine
view attributes reward functions to dopamine signaling injections induce a hyperlocomotion response in rodents,
in the nucleus accumbens (Wise 2004, Palmiter 2008). which has been ascribed to the drug’s dopamine-releasing
However, recent findings show that other dopamine actions (Salahpour et  al. 2008). Notably, ORX increases,
terminal fields are also components of the brain reward whilst testosterone restores the responsiveness to the
system, including the dorsal striatum, amygdala and locomotor effects of amphetamine in both rats and mice
frontal cortex (Wise 2004, Palmiter 2008). Following the (Purves-Tyson et al. 2015, Jardí et al. 2018). Overall, there
withdrawal of running wheels, mice selectively bred for is enough evidence to conclude that androgens influence
high wheel running showed a more pronounced increase the dopamine function in vivo. In addition, we recently
in striatal activation compared to controls (Rhodes et al. showed that systemic administration of the dopamine
2003). Also in male mice, the relative volume of the receptor 2 antagonist L-741,626 reduced by almost 50%
striatum predicted the levels of subsequent wheel running the distance ran by ORX+testosterone-treated mice,
activity on in vivo MRI (Cahill et al. 2015). whilst it had no effect on castrated mice receiving vehicle
Testosterone might act directly on male midbrain (Jardí et al. 2018). Further research is required to narrow
dopamine neurons by binding to their AR and/or ERs down the relevant androgen-responsive components of

Figure 3
Schematic overview of the proposed neuronal
mechanisms by which testosterone increases male
physical activity (see text). A, androgens; DA,
dopamine; E2, estradiol; MeA, medial amygdala;
NAcc, nucleus accumbens; SPA, spontaneous
physical activity; VMH, ventromedial
hypothalamus.

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access
Journal of F Jardí et al. Androgens and physical activity 238:1 R43
Endocrinology

the dopamine system that might be mediating the effects Nevertheless, other studies using larger cohorts and activity-
of testosterone on physical activity (Jardí et al. 2018). based questionnaires yielded conflicting observations (Amin
To summarize, the control of activity and metabolism et  al. 2000, Beutel et  al. 2005, Tajar et  al. 2012). Similarly,
are regulated in a coupled manner by estrogen-responsive there is no clear evidence from interventional studies that
hypothalamic pathways in females. The implication supports a causal effect of sex steroids on physical activity.
of similar mechanisms in males remains to be proven. Several double-blind, placebo-controlled clinical trials have
Extrahypothalamic inputs coming from brain regions examined the effects of testosterone replacement on both
related to emotional and rewarding processes might muscle as well as functional mobility in large samples of
modulate the homeostatic control exerted by the community-dwelling men with late-onset hypogonadism
hypothalamus. Testosterone might act on the MeA to (Emmelot-Vonk et  al. 2008, Srinivas-Shankar et  al. 2010,
increase physical activity in males following aromatization Snyder et al. 2016, Storer et al. 2016). Overall, testosterone
into E2 (Fig. 3). The dopamine system is another potential supplementation in hypogonadal men results in modest
candidate mediating the effects of testosterone on gains in muscle mass and strength, but the benefits on
activity, but further research is required to determine the their overall physical function are inconsistent (Emmelot-
mechanisms implicated (Fig. 3). Vonk et al. 2008, Srinivas-Shankar et al. 2010, Snyder et al.
2016, Storer et al. 2016). In addition, testosterone therapy
in men is hampered by its potential adverse effects on the
Human evidence cardiovascular system and the prostate (Basaria et al. 2010).
Therefore, the clinical meaningfulness of testosterone
After reviewing the available data, it is clear that androgens replacement in healthy elderly men with late-onset
regulate physical activity in male rodents, but in men, hypogonadism remains debated. Remarkably, treatment
it remains a question. The etiology of physical activity with testosterone for 6  months did not affect the self-
in humans appears to be much more complex than in reported levels of activity in a cohort of 274 community-
animals, being influenced by the social and cultural dwelling frail elderly men with low levels of testosterone
environments (Bauman et  al. 2012). Heritability studies (Srinivas-Shankar et  al. 2010). Smaller studies in similar
in twins and families suggest that our drive to engage in patient populations also showed a lack of testosterone effect
exercise is also determined by intrinsic biological processes on activity scores (Kenny et  al. 2001, 2010). Contrarily, a
(Simonen et al. 2002, Bouchard & Rankinen 2006, Stubbe positive association between serum testosterone levels and
et al. 2006). Nevertheless, the genetics of physical activity activity scores was found in a cohort of elderly men receiving
remain currently unknown, with only limited genotype– a combined treatment of testosterone and recombinant
phenotype associations found so far. A candidate gene human growth hormone (Sattler et al. 2011). The authors
study in postmenopausal women showed an association hypothesized that this association might reflect a central
between aromatase gene polymorphisms and time spent in action of testosterone on the individual’s mood (Sattler
physical activity (Salmen et  al. 2003). This association for et  al. 2011). To date, there is no real evidence supporting
the aromatase gene was replicated in a GWAS for leisure- the former premise, with the effects of testosterone on
time exercise behavior but only in one out of the two vitality and mood parameters in elderly men being unclear
populations of adult men included (De Moor et al. 2009). (Snyder et al. 1999, Srinivas-Shankar et al. 2010, Snyder et al.
Polymorphisms in the AR or ERα gene were not associated 2016). More importantly, all previous studies were based on
with the amount of physical activity per week in either activity questionnaires, which show limited reliability and
young or middle-aged men or women (Lorentzon et al. 1999, accuracy (Strath et al. 2013). Thus, caution should be taken
Salmén et  al. 2000, Okura et  al. 2003, Walsh et  al. 2005). when interpreting the data. In conclusion, it is premature to
Aging is associated with a decline in physical activity in both support or reject convincingly the implication of androgens
genders (Jones et al. 2011) as well as with a gradual reduction in physical activity in men.
in serum testosterone, especially its free fraction, in men
(Wu et al. 2008). So far, studies of the association between
late-onset hypogonadism and the age-related decline in Conclusion
activity have produced inconsistent results. A small cross-
sectional study in community-dwelling men showed an A wide body of preclinical evidence makes a strong case
association between low testosterone levels and a decrease for androgens as potential candidates contributing to
in pedometer-recorded step counts (Cobo et  al. 2017). the biological basis regulating male physical activity.

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain
Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access
Journal of F Jardí et al. Androgens and physical activity 238:1 R44
Endocrinology

Nonetheless, reliable human studies are lacking and


References
our present knowledge of the implicated processes is
Allen DL, Harrison BC, Maass A, Bell ML, Byrnes WC & Leinwand LA
very limited. This is explained in part by the fact that 2001 Cardiac and skeletal muscle adaptations to voluntary wheel
the interest in the biological factors determining activity running in the mouse. Journal of Applied Physiology 90 1900–1908.
levels is relatively recent. Measurement of physical activity (https://doi.org/10.1530/eje.0.1330361)
Almeida M, Laurent MR, Dubois V, Claessens F, O’Brien CA, Bouillon R,
in humans most often relies on self-reported instruments Vanderschueren D, Manolagas SC. 2017 Estrogens and androgens
due to their practicality and low cost. Nevertheless, these in skeletal physiology and pathophysiology. Physiological Reviews 97
measures have severe limitations in terms of reliability 135–187. (https://doi.org/10.1152/physrev.00033.2015)
American Physiological Association 2007 Exercise protocols using rats and
and validity, being subjected to several biases arising from mice. Resource Book for the Design of Animal Exercise Protocols, ch 3,
both the respondent and/or the investigator. In addition, pp 53–56. (https://doi.org/10.2460/ajvr.68.6.583)
the terminology as well as the analysis and interpretation Amin S, Zhang Y, Sawin CT, Evans SR, Hannan MT, Kiel DP, Wilson PWF &
Felson DT 2000 Association of hypogonadism and estradiol levels with
of the data differ markedly between studies. The current bone mineral density in elderly men from the Framingham study. Annals
challenge is to validate affordable objective measures that of Internal Medicine 133 951–963. (https://doi.org/200012190-00010 [pii])
can be implemented in large cohorts. In comparison, the Amory JK, Wang C, Swerdloff RS, Anawalt BD, Matsumoto AM,
Bremner WJ, Walker SE, Haberer LJ & Clark RV 2007 The effect of
mechanisms underlying the anabolic effects of androgens 5α-reductase inhibition with dutasteride and finasteride on semen
on muscle are better defined. Androgens increase muscle parameters and serum hormones in healthy men. Journal of Clinical
power in both young and elderly men, particularly when Endocrinology and Metabolism 92 1659–1665. (https://doi.org/10.1210/
jc.2006-2203)
combined with exercise. In male mice, myogenic AR Arnold AP 2009 The organizational-activational hypothesis as the
signaling drives in part the mild effects of testosterone foundation for a unified theory of sexual differentiation of all
stimulating appendicular muscle strength. Also in male mammalian tissues. Hormones and Behavior 55 570–578. (https://doi.
org/10.1016/j.yhbeh.2009.03.011)
mice, the modest and slow gains in muscle mass induced Atallah A, Mhaouty-Kodja S & Grange-Messent V 2017 Chronic depletion
by androgens contrast with their dramatic and fast actions of gonadal testosterone leads to blood-brain barrier dysfunction and
increasing wheel running. Taken together, preclinical inflammation in male mice. Journal of Cerebral Blood Flow and Metabolism
37 3161–3175. (https://doi.org/10.1177/0271678X16683961)
data do not support the notion that androgens stimulate Aubele T & Kritzer MF 2011 Gonadectomy and hormone replacement
physical activity primarily by favoring muscle function. affects in vivo basal extracellular dopamine levels in the prefrontal
On the other hand, the central nature of testosterone cortex but not motor cortex of adult male rats. Cerebral Cortex 21
222–232. (https://doi.org/10.1093/cercor/bhq083)
effects on activity is supported by the recent identification Aubele T & Kritzer MF 2012 Androgen influence on prefrontal dopamine
of sex-steroid-responsive neuronal circuitries modulating systems in adult male rats: localization of cognate intracellular
physical activity. Although data in males are still scarce, receptors in medial prefrontal projections to the ventral tegmental
area and effects of gonadectomy and hormone replacement on
the former findings indicate that brain aromatization is glutamate-stimulated. Cerebral Cortex 22 1799–1812. (https://doi.
required for the full action of testosterone on activity. org/10.1093/cercor/bhr258)
In conclusion, the CNS appears as the primary target of Axell A-M, MacLean HE, Plant DR, Harcourt LJ, Davis JA, Jimenez M,
Handelsman DJ, Lynch GS & Zajac JD 2006 Continuous testosterone
action for androgens stimulating physical activity, whilst administration prevents skeletal muscle atrophy and enhances
the multiple beneficial effects of testosterone on muscle resistance to fatigue in orchidectomized male mice. American Journal
and other peripheral organs do not drive the increase in of Physiology: Endocrinology and Metabolism 291 E506–E516. (https://
doi.org/10.1152/ajpendo.00058.2006)
activity but might contribute to the adaptation to exercise. Ayaz O & Howlett SE 2015 Testosterone modulates cardiac contraction
A better understanding of the underlying biological and calcium homeostasis: cellular and molecular mechanisms. Biology
mechanisms could have implications for the design of of Sex Differences 6 9. (https://doi.org/10.1186/s13293-015-0027-9)
Barone R, Pitruzzella A, Marino Gammazza A, Rappa F, Salerno M,
more effective interventions to promote physical activity. Barone F, Sangiorgi C, D’Amico D, Locorotondo N, Di Gaudio F, et al.
2017 Nandrolone decanoate interferes with testosterone biosynthesis
altering blood–testis barrier components. Journal of Cellular and
Molecular Medicine 21 1636–1647. (https://doi.org/10.1111/
Declaration of interest jcmm.13092)
The authors declare that there is no conflict of interest that could be Basaria S, Coviello AD, Travison TG, Storer TW, Farwell WR, Jette AM,
perceived as prejudicing the impartiality of this review. Eder R, Tennstedt S, Ulloor J, Zhang A, et al. 2010 Adverse events
associated with testosterone administration. New England Journal of
Medicine 363 109–122. (https://doi.org/10.1056/NEJMoa1000485)
Bauman AE, Reis RS, Sallis JF, Wells JC, Loos RJF & Martin BW 2012
Correlates of physical activity: why are some people physically active
Funding and others not? Lancet 380 258–271. (https://doi.org/10.1016/S0140-
This work was supported by the Research Foundation Flanders (FWO; grant 6736(12)60735-1)
G0D2217N) and the KU Leuven Research Council (grant GOA/15/017). F J is Berchtold NC, Kesslak JP, Pike CJ, Adlard PA & Cotman CW 2001
supported by a post-doctoral grant from FWO. Estrogen and exercise interact to regulate brain-derived neurotrophic

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access
Journal of F Jardí et al. Androgens and physical activity 238:1 R45
Endocrinology

factor mRNA and protein expression in the hippocampus. European Cahill LS, Steadman PE, Jones CE, Laliberté CL, Dazai J, Lerch JP,
Journal of Neuroscience 14 1992–2002. (https://doi.org/10.1046/j.0953- Stefanovic B & Sled JG 2015 MRI-detectable changes in mouse brain
816x.2001.01825.x) structure induced by voluntary exercise. NeuroImage 113 175–183.
Bergink EW, Janssen PSL, Turpun EW & Van Der Vies J 1985 Comparison (https://doi.org/10.1016/j.neuroimage.2015.03.036)
of the receptor binding properties of nandrolone and testosterone Callewaert F, Venken K, Ophoff J, De Gendt K, Torcasio A, van
under in vitro and in vivo conditions. Journal of Steroid Biochemistry Lenthe GH, Van Oosterwyck H, Boonen S, Bouillon R, Verhoeven G,
22 831–836. (https://doi.org/10.1016/0022-4731(85)90293-6) et al. 2009 Differential regulation of bone and body composition in
Beutel ME, Wiltink J, Hauck EW, Auch D, Behre HM, Brähler E & male mice with combined inactivation of androgen and estrogen
Weidner W 2005 Correlations between hormones, physical, and receptor-alpha. FASEB Journal  23 232–240. (https://doi.org/10.1096/
affective parameters in aging urologic outpatients. European Urology fj.08-113456)
47 749–755. (https://doi.org/10.1016/j.eururo.2005.02.015) Callewaert F, Boonen S & Vanderschueren D 2010 Sex steroids and the
Bhasin S, Travison TG, Storer TW, Lakshman K, Kaushik M, Mazer NA, male skeleton: a tale of two hormones. Trends in Endocrinology and
Ngyuen AH, Davda MN, Jara H, Aakil A, et al. 2012 Effect of Metabolism 21 89–95. (https://doi.org/10.1016/j.tem.2009.09.002)
testosterone supplementation with and without a dual 5α-reductase Caspersen CJ, Powell KE & Christenson GM 1985 Physical activity,
inhibitor on fat-free mass in men with suppressed testosterone exercise, and physical fitness: definitions and distinctions for health-
production: a randomized controlled trial. JAMA 307 931–939. related research. Public Health Reports 100 126–131. (https://doi.
(https://doi.org/10.1001/jama.2012.227) org/10.2307/20056429)
Bjørnebekk A, Walhovd KB, Jørstad ML, Due-Tønnessen P, Hullstein IR Chabert C, Bottelin P, Pison C & Dubouchaud H 2016 A low-cost system
& Fjell AM 2017 Structural brain imaging of long-term to easily measure spontaneous physical activity in rodents. Journal
anabolic-androgenic steroid users and nonusing weightlifters. of Applied Physiology 120 1097–1103. (https://doi.org/10.1152/
Biological Psychiatry 82 294–302. (https://doi.org/10.1016/j. japplphysiol.00888.2015)
biopsych.2016.06.017) Chambon C, Duteil D, Vignaud A, Ferry A, Messaddeq N, Malivindi R,
Blattner MS & Mahoney MM 2012 Circadian parameters are altered Kato S, Chambon P & Metzger D 2010 Myocytic androgen receptor
in two strains of mice with transgenic modifications of estrogen controls the strength but not the mass of limb muscles. PNAS 107
receptor subtype 1. Genes, Brain and Behavior 11 828–836. (https://doi. 14327–14332. (https://doi.org/10.1073/pnas.1009536107)
org/10.1111/j.1601-183X.2012.00831.x) Chen CV, Brummet JL, Jordan CL & Breedlove SM 2016 Down, but
Blattner MS & Mahoney MM 2015 Changes in estrogen receptor signaling not out: partial elimination of androgen receptors in the male
alters the timekeeping system in male mice. Behavioural Brain Research mouse brain does not affect androgenic regulation of anxiety or
294 43–49. (https://doi.org/10.1016/j.bbr.2015.07.060) HPA activity. Endocrinology 157 764–773. (https://doi.org/10.1210/
Blizard DA 1983 Sex differences in running-wheel behaviour in the en.2015-1417)
rat: the inductive and activational effects of gonadal hormones. Chisamore MJ, Gentile MA, Dillon GM, Baran M, Gambone C, Riley S,
Animal Behaviour 31 378–384. (https://doi.org/10.1016/S0003- Schmidt A, Flores O, Wilkinson H & Alves SE 2016 A novel selective
3472(83)80056-6) androgen receptor modulator (SARM) MK-4541 exerts anti-
Boecker H, Sprenger T, Spilker ME, Henriksen G, Koppenhoefer M, androgenic activity in the prostate cancer xenograft R–3327G and
Wagner KJ, Valet M, Berthele A & Tolle TR 2008 The runner’s high: anabolic activity on skeletal muscle mass & function in castrated
opioidergic mechanisms in the human brain. Cerebral Cortex 18 mice. Journal of Steroid Biochemistry and Molecular Biology 163 88–97.
2523–2531. (https://doi.org/10.1093/cercor/bhn013) (https://doi.org/10.1016/j.jsbmb.2016.04.007)
Borst SE, Conover CF, Carter CS, Gregory CM, Marzetti E, Cobo G, Gallar P, Di Gioia C, García Lacalle C, Camacho R, Rodriguez I,
Leeuwenburgh C, Vandenborne K & Wronski TJ 2007 Anabolic Ortega O, Mon C, Vigil A, Lindholm B, et al. 2017 Hypogonadism
effects of testosterone are preserved during inhibition of 5-reductase. associated with muscle atrophy, physical inactivity and ESA
AJP: Endocrinology and Metabolism 293 E507–E514. (https://doi. hyporesponsiveness in men undergoing haemodialysis. Nefrologia :
org/10.1152/ajpendo.00130.2007) Publicacion Oficial de La Sociedad Espanola Nefrologia 37 54–60.
Bouchard C & Rankinen T 2006 Are people physically inactive because of (https://doi.org/10.1016/j.nefro.2016.04.009)
their genes? President’s Council on Physical Fitness and Sports Research Cooke PS, Nanjappa MK, Ko C, Prins GS & Hess RA 2017 Estrogens in
Digest 7 1–8. male physiology. Physiological Reviews 97 995–1043. (https://doi.
Bowen RS, Ferguson DP & Lightfoot JT 2011 Effects of aromatase inhibition org/10.1152/physrev.00018.2016)
on the physical activity levels of male mice. Journal of Steroids and Correa SM, Newstrom DW, Warne JP, Flandin P, Cheung CC, Lin-
Hormonal Science 1 1–7. (https://doi.org/10.4172/2157-7536.S1-001) Moore AT, Pierce AA, Xu AW, Rubenstein JL & Ingraham HA 2015
Bowen RS, Knab AM, Hamilton AT, McCall JR, Moore-Harrison TL & An estrogen-responsive module in the ventromedial hypothalamus
Lightfoot JT 2012 Effects of supraphysiological doses of sex steroids selectively drives sex-specific activity in females. Cell Reports 10
on wheel running activity in mice. Journal of Steroids and Hormonal 62–74. (https://doi.org/10.1016/j.celrep.2014.12.011)
Science 3 110. (https://doi.org/10.4172/2157-7536.1000110) Cozzoli A, Capogrosso RF, Sblendorio VT, Dinardo MM, Jagerschmidt C,
Brock O, Baum MJ & Bakker J 2011 The development of female sexual Namour F, Camerino GM & De Luca A 2013 GLPG0492, a novel
behavior requires prepubertal estradiol. Journal of Neuroscience 31 selective androgen receptor modulator, improves muscle performance
5574–5578. (https://doi.org/10.1523/JNEUROSCI.0209-11.2011) in the exercised-mdx mouse model of muscular dystrophy.
Brockman R, Bunick D & Mahoney MM 2011 Estradiol deficiency Pharmacological Research 72 9–24. (https://doi.org/10.1016/j.
during development modulates the expression of circadian and daily phrs.2013.03.003)
rhythms in male and female aromatase knockout mice. Hormones and Curzon P, Zhang M, Radek RJ & Fox GB 2009 The behavioral assessment
Behavior 60 439–447. (https://doi.org/10.1016/j.yhbeh.2011.07.011) of sensorimotor processes in the mouse: acoustic startle, sensory
Broida J & Svare B 1984 Sex differences in the activity of mice: gating, locomotor activity, rotarod, and beam walking. In Methods of
modulation by postnatal gonadal hormones. Hormones and Behavior Behavior Analysis in Neuroscience, pp 1–28. Boca Raton, FL, USA: Taylor
18 65–78. (https://doi.org/10.1016/0018-506X(84)90051-5) & Francis.
Butler MP, Karatsoreos IN, Le Sauter J & Silver R 2012 Dose-dependent Daan S, Damassa D, Pittendrigh CS & Smith ER 1975 An effect of
effects of androgens on the circadian timing system and its response castration and testosterone replacement on a circadian pacemaker in
to light. Endocrinology 153 2344–2352. (https://doi.org/10.1210/ mice (Mus musculus). PNAS 72 3744–3747. (https://doi.org/10.1073/
en.2011-1842) pnas.72.9.3744)

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain
Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access
Journal of F Jardí et al. Androgens and physical activity 238:1 R46
Endocrinology

Dark J & Zucker I 1984 Gonadal and photoperiodic control of seasonal cardiac performance and oxidative stress in orchidectomized rats. Acta
body weight changes in male voles. American Journal of Physiology 247 Physiologica 209 136–147. (https://doi.org/10.1111/apha.12158)
R84–R88. Emmelot-Vonk MH, Verhaar HJJ, Nakhai Pour HR, Aleman A,
Davey RA, Clarke MV, Russell PK, Rana K, Seto J, Roeszler KN, How JMY, Lock TMTW, Bosch JLHR, Grobbee DE & Van Der Schouw YT 2008
Chia LY, North K & Zajac JD 2017 Androgen action via the androgen Effect of testosterone supplementation on functional mobility,
receptor in neurons within the brain positively regulates muscle mass cognition, and other parameters in older men: a randomized
in male mice. Endocrinology 158 3684–3695. (https://doi.org/10.1210/ controlled trial. JAMA 299 39–52. (https://doi.org/10.1001/
en.2017-00470) jama.2007.51)
Deacon RMJ 2013 Measuring motor coordination in mice. Journal of Eriksson M, Rasmussen F & Tynelius P 2006 Genetic factors in physical
Visualized Experiments 75 e2609. (https://doi.org/10.3791/2609) activity and the equal environment assumption – the Swedish
Delbeck M, Golz S, Vonk R, Janssen W, Hucho T, Isensee J, Schäfer S young male twins study. Behavior Genetics 36 238–247. (https://doi.
& Otto C 2011 Impaired left-ventricular cardiac function in male org/10.1007/s10519-005-9018-7)
GPR30-deficient mice. Molecular Medicine Reports 4 37–40. (https://doi. Fan WQ, Yanase T, Nomura M, Okabe T, Goto K, Sato T, Kawano H,
org/10.3892/mmr.2010.402) Kato S & Nawata H 2005 Androgen receptor null male mice
De Lorme KC & Sisk CL 2016 The organizational effects of pubertal develop late-onset obesity caused by decreased energy expenditure
testosterone on sexual proficiency in adult male Syrian hamsters. and lipolytic activity but show normal insulin sensitivity with
Physiology and Behavior 165 273–277. (https://doi.org/10.1016/j. high adiponectin secretion. Diabetes 54 1000–1008. (https://doi.
physbeh.2016.08.008) org/10.2337/diabetes.54.4.1000)
De Moor MHM, Liu Y-J, Boomsma DI, Li J, Hamilton JJ, Hottenga J, Fernandes MFA, Matthys D, Hryhorczuk C, Sharma S, Mogra S, Alquier T
Levy S, Liu X, Pei Y, Posthuma D, et al. 2009 Genome-wide & Fulton S 2015 Leptin suppresses the rewarding effects of running
association study of exercise behavior in Dutch and American adults. via STAT3 signaling in dopamine neurons. Cell Metabolism 22
Medicine and Science in Sports and Exercise 41 1887–1895. (https://doi. 741–749. (https://doi.org/10.1016/j.cmet.2015.08.003)
org/10.1249/MSS.0b013e3181a2f646) Ferry A, Schuh M, Parlakian A, Mgrditchian T, Valnaud N, Joanne P,
De Naeyer H, Lamon S, Russell AP, Everaert I, De Spaey A, Vanheel B, Butler-Browne G, Agbulut O & Metzger D 2014 Myofiber androgen
Taes Y & Derave W 2014 Androgenic and estrogenic regulation of receptor promotes maximal mechanical overload-induced muscle
Atrogin-1, MuRF1 and myostatin expression in different muscle types hypertrophy and fiber type transition in male mice. Endocrinology 155
of male mice. European Journal of Applied Physiology 114 751–761. 4739–4748. (https://doi.org/10.1210/en.2014-1195)
(https://doi.org/10.1007/s00421-013-2800-y) Fisher CR, Graves KH, Parlow AF & Simpson ER 1998 Characterization
De Visser L, Van Den Bos R & Spruijt BM 2005 Automated home cage of mice deficient in aromatase (ArKO) because of targeted disruption
observations as a tool to measure the effects of wheel running on cage of the cyp19 gene. Medical Sciences 95 6965–6970. (https://doi.
floor locomotion. Behavioural Brain Research 160 382–388. (https:// org/10.1073/pnas.95.12.6965)
doi.org/10.1016/j.bbr.2004.12.004) Fragkaki AG, Angelis YS, Koupparis M, Tsantili-Kakoulidou A, Kokotos G
De Vries GJ, Rissman EF, Simerly RB, Yang L-Y, Scordalakes EM, Auger CJ, & Georgakopoulos C 2009 Structural characteristics of anabolic
Swain A, Lovell-Badge R, Burgoyne PS & Arnold AP 2002 A model androgenic steroids contributing to binding to the androgen
system for study of sex chromosome effects on sexually dimorphic receptor and to their anabolic and androgenic activities. Applied
neural and behavioral traits. Journal of Neuroscience 22 9005–9014. modifications in the steroidal structure. Steroids 74 172–197. (https://
(https://doi.org/22/20/9005) doi.org/10.1016/j.steroids.2008.10.016)
Dubois V, Laurent M, Boonen S, Vanderschueren D & Claessens F Frye CA, Van Keuren KR & Erskine MS 1996 Behavioral effects of
2012 Androgens and skeletal muscle: cellular and molecular action 3α-androstanediol I: modulation of sexual receptivity and promotion
mechanisms underlying the anabolic actions. Cellular and Molecular Life of GABA-stimulated chloride flux. Behavioural Brain Research 79
Sciences 69 1651–1667. (https://doi.org/10.1007/s00018-011-0883-3) 109–118. (https://doi.org/10.1016/S0166-4328(96)00004-6)
Dubois V, Laurent MR, Sinnesael M, Cielen N, Helsen C, Clinckemalie L, Frye CA, Koonce CJ, Edinger KL, Osborne DM & Walf AA 2008a
Spans L, Gayan-Ramirez G, Deldicque L, Hespel P, et al. 2014 A Androgens with activity at estrogen receptor beta have anxiolytic
satellite cell-specific knockout of the androgen receptor reveals and cognitive-enhancing effects in male rats and mice. Hormones and
myostatin as a direct androgen target in skeletal muscle. FASEB Journal Behavior 54 726–734. (https://doi.org/10.1016/j.yhbeh.2008.07.013)
28 2979–2994. (https://doi.org/10.1096/fj.14-249748) Frye CA, Edinger K & Sumida K 2008b Androgen administration to aged
Dubois V, Simitsidellis I, Laurent MR, Jardi F, Saunders PTK, male mice increases anti-anxiety behavior and enhances cognitive
Vanderschueren D & Claessens F 2015 Enobosarm (GTx-024) performance. Neuropsychopharmacology 33 1049–1061. (https://doi.
modulates adult skeletal muscle mass independently of the androgen org/10.1038/sj.npp.1301498)
receptor in the satellite cell lineage. Endocrinology 156 4522–4533. Gans HM 1927 Studies on vigor. Effect of fractional castration on the
(https://doi.org/10.1210/en.2015-1479) voluntary activity of male albino rats. Endocrinology 11 145–148.
Dubois V, Laurent MR, Jardi F, Antonio L, Lemaire K, Goyvaerts L, (https://doi.org/10.1210/endo-11-2-145)
Deldicque L, Carmeliet G, Decallonne B, Vanderschueren D, Garland T, Schutz H, Chappell MA, Keeney BK, Meek TH, Copes LE,
et al. 2016 Androgen deficiency exacerbates high-fat diet-induced Acosta W, Drenowatz C, Maciel RC, van Dijk G, et al. 2011 The
metabolic alterations in male mice. Endocrinology 157 648–665. biological control of voluntary exercise, spontaneous physical activity
(https://doi.org/10.1210/en.2015-1713) and daily energy expenditure in relation to obesity: human and
Dworatzek E, Mahmoodzadeh S, Schubert C, Westphal C, Leber J, rodent perspectives. Journal of Experimental Biology 214 206–229.
Kusch A, Kararigas G, Fliegner D, Moulin M, Ventura-Clapier R, et al. (https://doi.org/10.1242/jeb.048397)
2014 Sex differences in exercise-induced physiological myocardial Gentry RT & Wade GN 1976 Sex differences in sensitivity of food intake,
hypertrophy are modulated by oestrogen receptor beta. Cardiovascular body weight, and running- wheel activity to ovarian steroids in
Research 102 418–428. (https://doi.org/10.1093/cvr/cvu065) rats. Journal of Comparative and Physiological Psychology 90 747–754.
Eikelboom R & Mills R 1988 A microanalysis of wheel running in male (https://doi.org/10.1037/h0077246)
and female rats. Physiology and Behavior 43 625–630. (https://doi. Georgieva KN & Boyadjiev NP 2004 Effects of nandrolone decanoate
org/10.1016/0031-9384(88)90217-X) on V̇O2max, running economy, and endurance in rats. Medicine
Eleawa SM, Sakr HF, Hussein AM, Assiri AS, Bayoumy NMK & and Science in Sports and Exercise 36 1336–1341. (https://doi.
Alkhateeb M 2013 Effect of testosterone replacement therapy on org/10.1249/01.MSS.0000135781.42515.17)

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access
Journal of F Jardí et al. Androgens and physical activity 238:1 R47
Endocrinology

Gofflot F, Chartoire N, Vasseur L, Heikkinen S, Dembele D, Le Merrer J & Jardí F, Laurent MR, Khalil R, Kim N, Dubois V, Deboel L, Schollaert D,
Auwerx J 2007 Systematic gene expression mapping clusters nuclear Carmeliet G, Decallonne B, Claessens F, et al. 2017b Deletion of
receptors according to their function in the brain. Cell 131 405–418. androgen receptor in neurons accelerates the age-related loss of
(https://doi.org/10.1016/j.cell.2007.09.012) cortical thickness in male mice. Journal of Bone and Mineral Research
Guo W, Wong S, Li M, Liang W, Liesa M, Serra C, Jasuja R, Bartke A, 32 1152.
Kirkland JL, Shirihai O, et al. 2012 Testosterone plus low-intensity Jardí F, Laurent MR, Kim N, Khalil R, De Bundel D, Van Eeckhaut A,
physical training in late life improves functional performance, Van Helleputte L, Deboel L, Dubois V, Schollaert D, et al. 2018
skeletal muscle mitochondrial biogenesis, and mitochondrial quality Testosterone boosts physical activity in male mice via dopaminergic
control in male mice. PLoS ONE 7 e51180. (https://doi.org/10.1371/ pathways. Scientific Reports 8 957.
journal.pone.0051180) Jayaraman A, Lent-Schochet D & Pike CJ 2014 Diet-induced obesity
Hallal PC, Andersen LB, Bull FC, Guthold R, Haskell W, Ekelund U, and low testosterone increase neuroinflammation and impair
Alkandari JR, Bauman AE, Blair SN, Brownson RC, et al. 2012 neural function. Journal of Neuroinflammation 11 162. (https://doi.
Global physical activity levels: surveillance progress, pitfalls, and org/10.1186/s12974-014-0162-y)
prospects. Lancet 380 247–257. (https://doi.org/10.1016/S0140- Jechura TJ, Walsh JM & Lee TM 2000 Testicular hormones modulate
6736(12)60646-1) circadian rhythms of the diurnal rodent, Octodon degus. Hormones
Hamrick MW, Ding K-H, Pennington C, Chao YJ, Wu Y-D, Howard B, and Behavior 38 243–249. (https://doi.org/10.1006/hbeh.2000.1624)
Immel D, Borlongan C, McNeil PL, Bollag WB, et al. 2006 Age-related Jones ME, Thorburn AW, Britt KL, Hewitt KN, Wreford NG, Proietto J,
loss of muscle mass and bone strength in mice is associated with Oz OK, Leury BJ, Robertson KM, Yao S, et al. 2000 Aromatase-deficient
a decline in physical activity and serum leptin. Bone 39 845–853. (ArKO) mice have a phenotype of increased adiposity. PNAS 97
(https://doi.org/10.1016/j.bone.2006.04.011) 12735–12740. (https://doi.org/10.1073/pnas.97.23.12735)
Harno E, Cottrell EC & White A 2013 Metabolic pitfalls of CNS cre-based Jones H, Millward P & Buraimo B 2011 Adult participation in sport.
technology. Cell Metabolism 18 21–28. (https://doi.org/10.1016/j. Analysis of the taking part survey. London, UK: Department for
cmet.2013.05.019) Digital, Culture, Media & Sport. (available at: https://www.gov.uk/
Heath GW, Parra DC, Sarmiento OL, Andersen LB, Owen N, Goenka S, government/publications/adult-participation-in-sport-analysis-of-the-
Montes F, Brownson RC, Alkandari JR, Bauman AE, et al. 2012 taking-part-survey)
Evidence-based intervention in physical activity: lessons from around Juntti SA, Tollkuhn J, Wu MV, Fraser EJ, Soderborg T, Tan S, Honda SI,
the world. Lancet 380 272–281. (https://doi.org/10.1016/S0140- Harada N & Shah NM 2010 The androgen receptor governs the
6736(12)60816-2) execution, but not programming, of male sexual and territorial
Heine PA, Taylor JA, Iwamoto GA, Lubahn DB & Cooke PS 2000 behaviors. Neuron 66 260–272. (https://doi.org/10.1016/j.
Increased adipose tissue in male and female estrogen receptor-alpha neuron.2010.03.024)
knockout mice. PNAS 97 12729–12734. (https://doi.org/10.1073/ Karatsoreos IN, Wang A, Sasanian J & Silver R 2007 A role for androgens
pnas.97.23.12729) in regulating circadian behavior and the suprachiasmatic nucleus.
Hill RA, McInnes KJ, Gong ECH, Jones MEE, Simpson ER & Boon WC Endocrinology 148 5487–5495. (https://doi.org/10.1210/en.2007-0775)
2007 Estrogen deficient male mice develop compulsive behavior. Kastenberger I & Schwarzer C 2014 GPER1 (GPR30) knockout mice
Biological Psychiatry 61 359–366. (https://doi.org/10.1016/j. display reduced anxiety and altered stress response in a sex and
biopsych.2006.01.012) paradigm dependent manner. Hormones and Behavior 66 628–636.
Hourdé C, Jagerschmidt C, Clément-Lacroix P, Vignaud A, Ammann P, (https://doi.org/10.1016/j.yhbeh.2014.09.001)
Butler-Browne GS & Ferry A 2009 Androgen replacement therapy Kelly DM & Jones TH 2013 Testosterone: a metabolic hormone in health
improves function in male rat muscles independently of hypertrophy and disease. Journal of Endocrinology 217 R25–R45. (https://doi.
and activation of the Akt/mTOR pathway. Acta Physiologica 195 org/10.1530/JOE-12-0455)
471–482. (https://doi.org/10.1111/j.1748-1716.2008.01902.x) Kelly DM, Akhtar S, Sellers DJ, Muraleedharan V, Channer KS & Jones TH
Ibebunjo C, Eash JK, Li C, Ma Q & Glass DJ 2011 Voluntary running, 2016 Testosterone differentially regulates targets of lipid and glucose
skeletal muscle gene expression, and signaling inversely regulated metabolism in liver, muscle and adipose tissues of the testicular
by orchidectomy and testosterone replacement. American Journal of feminised mouse. Endocrine 54 504–515. (https://doi.org/10.1007/
Physiology-Endocrinology and Metabolism 300 E327–E340. (https://doi. s12020-016-1019-1)
org/10.1152/ajpendo.00402.2010\r10.1152/ajpendo.00402.2010) Kenny AM, Prestwood KM, Gruman CA, Marcello KM & Raisz LG 2001
Iwahana E, Karatsoreos I, Shibata S & Silver R 2008 Gonadectomy reveals Effects of transdermal testosterone on bone and muscle in older men
sex differences in circadian rhythms and suprachiasmatic nucleus with low bioavailable testosterone levels. Journal of Gerontology 56A
androgen receptors in mice. Hormones and Behavior 53 422–430. M266–M277. (https://doi.org/10.1111/j.1532-5415.2010.02865.x)
(https://doi.org/10.1016/j.yhbeh.2007.11.014) Kenny AM, Kleppinger A, Annis K, Rathier M, Browner B, Judge JO
Jakacka M, Ito M, Martinson F, Ishikawa T, Lee EJ & Jameson JL 2002 & McGee D 2010 Effects of transdermal testosterone on bone
An estrogen receptor (ER)α deoxyribonucleic acid-binding domain and muscle in older men with low bioavailable testosterone
knock-in mutation provides evidence for nonclassical ER pathway levels, low bone mass, and physical frailty. Journal of the American
signaling in vivo. Molecular Endocrinology 16 2188–2201. (https://doi. Geriatrics Society 58 1134–1143. (https://doi.org/10.1111/j.1532-
org/10.1210/me.2001-0174) 5415.2010.02865.x)
Jänne M, Deol HK, Power SGA, Yee S & Hammond GL 1998 Human Keshavarzi S, Sullivan RKP, Ianno DJ & Sah P 2014 Functional
sex hormone-binding globulin gene expression in transgenic mice. properties and projections of neurons in the medial amygdala.
Molecular Endocrinology 12 123–136. (https://doi.org/10.1210/ Journal of Neuroscience 34 8699–8715. (https://doi.org/10.1523/
mend.12.1.0050) JNEUROSCI.1176-14.2014)
Jardí F, Laurent MR, Dubois V, Khalil R, Deboel L, Schollaert D, Van Khasnavis S, Ghosh A, Roy A & Pahan K 2013 Castration induces
Den Bosch L, Decallonne B, Carmeliet G, Claessens F, et al. 2017a A parkinson disease pathologies in young male mice via inducible
shortened tamoxifen induction scheme to induce CreER recombinase nitric-oxide synthase. Journal of Biological Chemistry 288
without side effects on the male mouse skeleton. Molecular and 20843–20855. (https://doi.org/10.1074/jbc.M112.443556)
Cellular Endocrinology 452 57–63. (https://doi.org/10.1016/j. Kim J, Min H, Oh S, Kim Y, Lee AH & Park T 2014 Joint identification
mce.2017.05.012) of genetic variants for physical activity in Korean population.

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain
Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access
Journal of F Jardí et al. Androgens and physical activity 238:1 R48
Endocrinology

International Journal of Molecular Sciences 15 12407–12421. (https:// Lin H-Y, Yu I-C, Wang R-S, Chen Y-T, Liu N-C, Altuwaijri S, Hsu C-
doi.org/10.3390/ijms150712407) L, Ma W-L, Jokinen J, Sparks JD, et al. 2008 Increased hepatic
Kim J-H, Han G-C, Seo J-Y, Park I, Park W, Jeong H-W, Lee SH, Bae S, steatosis and insulin resistance in mice lacking hepatic androgen
Seong J, Yum M-K, et al. 2016 Sex hormones establish a reserve pool receptor. Hepatology 47 1924–1935. (https://doi.org/10.1002/
of adult muscle stem cells. Nature Cell Biology 18 930–940. (https:// hep.22252)
doi.org/10.1038/ncb3401) Locklear MN, Michealos M, Collins WF & Kritzer MF 2017 Gonadectomy
Kohl HW, Craig CL, Lambert EV, Inoue S, Alkandari JR, Leetongin G & but not biological sex affects burst-firing in dopamine neurons
Kahlmeier S 2012 The pandemic of physical inactivity: global action of the ventral tegmental area and in prefrontal cortical neurons
for public health. Lancet 380 294–305. (https://doi.org/10.1016/ projecting to the ventral tegmentum in adult rats. European Journal of
S0140-6736(12)60898-8) Neuroscience 45 106–120. (https://doi.org/10.1111/ejn.13380)
Kritzer MF 2003 Long-term gonadectomy affects the density of Loos RJF, Rankinen T, Tremblay A, Pérusse L, Chagnon Y & Bouchard C
tyrosine hydroxylase- but not dopamine-β-hydroxylase-, choline 2005 Melanocortin-4 receptor gene and physical activity in the
acetyltransferase- or serotonin-immunoreactive axons in the medial Québec Family Study. International Journal of Obesity 29 420–428.
prefrontal cortices of adult male rats. Cerebral Cortex 13 282–296. (https://doi.org/10.1038/sj.ijo.0802869)
(https://doi.org/10.1093/cercor/13.3.282) Lorentzon M, Lorentzon R, Bäckström T & Nordström P 1999 Estrogen
Kritzer MF & Creutz LM 2008 Region and sex differences in constituent receptor gene polymorphism, but not estradiol levels, is related
dopamine neurons and immunoreactivity for intracellular estrogen to bone density in healthy adolescent boys: a cross-sectional and
and androgen receptors in mesocortical projections in rats. longitudinal study. Journal of Clinical Endocrinology and Metabolism 84
Journal of Neuroscience 28 9525–9535. (https://doi.org/10.1523/ 4597–4601.
JNEUROSCI.2637-08.2008) MacLean HE, Chiu WSM, Notini AJ, Axell A-M, Davey RA, McManus JF,
Kritzer MF, McLaughlin PJ, Smirlis T & Robinson JK 2001 Gonadectomy Ma C, Plant DR, Lynch GS & Zajac JD 2008 Impaired skeletal muscle
impairs T-maze acquisition in adult male rats. Hormones and Behavior development and function in male, but not female, genomic
39 167–174. (https://doi.org/10.1006/hbeh.2001.1645) androgen receptor knockout mice. FASEB Journal 22 2676–2689.
Kuljis DA, Loh DH, Truong D, Vosko AM, Ong ML, McClusky R, (https://doi.org/10.1096/fj.08-105726)
Arnold AP & Colwell CS 2013 Gonadal- and sex-chromosome- Magnaghi V, Cavarretta I, Zucchi I, Susani L, Rupprecht R, Hermann B,
dependent sex differences in the circadian system. Endocrinology 154 Martini L & Melcangi RC 1999 Po gene expression is modulated
1501–1512. (https://doi.org/10.1210/en.2012-1921) by androgens in the sciatic nerve of adult male rats. Molecular
Langer G, Bader B, Meoli L, Isensee J, Delbeck M, Noppinger PR & Otto C Brain Research 70 36–44. (https://doi.org/10.1016/S0169-
2010 A critical review of fundamental controversies in the field of 328X(99)00124-2)
GPR30 research. Steroids 75 603–610. (https://doi.org/10.1016/j. Magnaghi V, Ballabio M, Gonzalez LC, Leonelli E, Motta M &
steroids.2009.12.006) Melcangi RC 2004 The synthesis of glycoprotein Po and peripheral
Laurent MR, Helsen C, Antonio L, Schollaert D, Joniau S, Vos MJ, myelin protein 22 in sciatic nerve of male rats is modulated by
Decallonne B, Hammond GL, Vanderschueren D & Claessens F testosterone metabolites. Molecular Brain Research 126 67–73. (https://
2016a Effects of sex hormone-binding globulin (SHBG) on androgen doi.org/10.1016/j.molbrainres.2004.03.009)
bioactivity in vitro. Molecular and Cellular Endocrinology 437 280–291. Mahfouz A, Lelieveldt BPF, Grefhorst A, van Weert LTCM, Mol IM,
(https://doi.org/10.1016/j.mce.2016.08.041) Sips HCM, van den Heuvel JK, Datson NA, Visser JA, Reinders MJT,
Laurent MR, Hammond GL, Blokland M, Jardí F, Antonio L, Dubois V, et al. 2016 Genome-wide coexpression of steroid receptors in the
Khalil R, Sterk SS, Gielen E, Decallonne B, et al. 2016b Sex hormone- mouse brain: identifying signaling pathways and functionally
binding globulin regulation of androgen bioactivity in vivo: coordinated regions. PNAS 113 2738–2743. (https://doi.org/10.1073/
validation of the free hormone hypothesis. Scientific Reports 6 35539. pnas.1520376113)
(https://doi.org/10.1038/srep35539) Mao J-H, Langley SA, Huang Y, Hang M, Bouchard KE, Celniker SE,
Lee IM, Shiroma EJ, Lobelo F, Puska P, Blair SN, Katzmarzyk PT, Brown JB, Jansson JK, Karpen GH & Snijders AM 2015 Identification
Alkandari JR, Andersen LB, Bauman AE, Brownson RC, et al. 2012 of genetic factors that modify motor performance and body weight
Effect of physical inactivity on major non-communicable diseases using Collaborative Cross mice. Scientific Reports 5 16247. (https://doi.
worldwide: an analysis of burden of disease and life expectancy. Lancet org/10.1038/srep16247)
380 219–229. (https://doi.org/10.1016/S0140-6736(12)61031-9) Marie-Luce C, Raskin K, Bolborea M, Monin M, Picot M & Mhaouty-
Lein ES, Hawrylycz MJ, Ao N, Ayres M, Bensinger A, Bernard A, Boe AF, Kodja S 2013 Effects of neural androgen receptor disruption on
Boguski MS, Brockway KS, Byrnes EJ, et al. 2007 Genome-wide atlas aggressive behavior, arginine vasopressin and galanin systems in
of gene expression in the adult mouse brain. Nature 445 168–176. the bed nucleus of stria terminalis and lateral septum. General and
(https://doi.org/10.1038/nature05453) Comparative Endocrinology 188 218–225. (https://doi.org/10.1016/j.
Lenard NR & Berthoud H-R 2008 Central and peripheral regulation of ygcen.2013.03.031)
food intake and physical activity: pathways and genes. Obesity 16 McCarthy MM, Schlenker EH & Pfaff DW 1993 Enduring consequences
S11–S22. (https://doi.org/10.1038/oby.2008.511) of neonatal treatment with antisense oligodeoxynucleotides to
Lenz KM & McCarthy MM 2010 Organized for sex – steroid hormones estrogen receptor messenger ribonucleic acid on sexual differentiation
and the developing hypothalamus. European Journal of Neuroscience 32 of rat brain. Endocrinology 133 433–439. (https://doi.org/10.1210/
2096–2104. (https://doi.org/10.1111/j.1460-9568.2010.07511.x) endo.133.2.8344188)
Lett BT, Grant VL & Koh MT 2001 Naloxone attenuates the conditioned McDermott JL, Kreutzberg JD, Liu B & Dluzen DE 1994 Effects of
place preference induced by wheel running in rats. Physiology estrogen treatment on sensorimotor task performance and brain
and Behavior 72 355–358. (https://doi.org/10.1016/S0031- dopamine concentrations in gonadectomized male and female CD-1
9384(00)00427-3) mice. Hormones and Behavior 28 16–28. (https://doi.org/10.1006/
Levine JA 1999 Role of nonexercise activity thermogenesis in resistance hbeh.1994.1002)
to fat gain in humans. Science 283 212–214. (https://doi.org/10.1126/ McGinnis MY, Lumia AR, Tetel MJ, Molenda-Figueira HA & Possidente B
science.283.5399.212) 2007 Effects of anabolic androgenic steroids on the development and
Lightfoot JT 2008 Sex hormones’ regulation of rodent physical activity: a expression of running wheel activity and circadian rhythms in male
review. International Journal of Biological Sciences 4 126–132. (https:// rats. Physiology and Behavior 92 1010–1018. (https://doi.org/10.1016/j.
doi.org/10.7150/ijbs.4.126) physbeh.2007.07.010)

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access
Journal of F Jardí et al. Androgens and physical activity 238:1 R49
Endocrinology

McInnes KJ, Smith LB, Hunger NI, Saunders PTK, Andrew R & Walker BR Ophoff J, Callewaert F, Venken K, De Gendt K, Ohlsson C, Gayan-
2012 Deletion of the androgen receptor in adipose tissue in male mice Ramirez G, Decramer M, Boonen S, Bouillon R, Verhoeven G, et al.
elevates retinol binding protein 4 and reveals independent effects on 2009a Physical activity in the androgen receptor knockout mouse:
visceral fat mass and on glucose homeostasis. Diabetes 61 1072–1081. evidence for reversal of androgen deficiency on cancellous bone.
(https://doi.org/10.2337/db11-1136) Biochemical and Biophysical Research Communications 378 139–144.
McNamara KM, Harwood DT, Simanainen U, Walters KA, Jimenez M & (https://doi.org/10.1016/j.bbrc.2008.11.016)
Handelsman DJ 2010 Measurement of sex steroids in murine blood Ophoff J, Van Proeyen K, Callewaert F, De Gendt K, De Bock K, Vanden
and reproductive tissues by liquid chromatography-tandem mass Bosch A, Verhoeven G, Hespel P & Vanderschueren D 2009b
spectrometry. Journal of Steroid Biochemistry and Molecular Biology 121 Androgen signaling in myocytes contributes to the maintenance of
611–618. (https://doi.org/10.1016/j.jsbmb.2010.02.001) muscle mass and fiber type regulation but not to muscle strength
Mhillaj E, Morgese MG, Tucci P, Bove M, Schiavone S & Trabace L 2015 or fatigue. Endocrinology 150 3558–3566. (https://doi.org/10.1210/
Effects of anabolic-androgens on brain reward function. Frontiers in en.2008-1509)
Neuroscience 9 295. (https://doi.org/10.3389/fnins.2015.00295) Oz K, Zerwekh JE, Fisher C, Graves K, Nanu L, Millsaps R & Simpson ER
Model Z, Butler MP, LeSauter J & Silver R 2015 Suprachiasmatic nucleus 2000 Bone has a sexually dimorphic response to aromatase deficiency.
as the site of androgen action on circadian rhythms. Hormones and Journal of Bone and Mineral Research 15 507–514. (https://doi.
Behavior 73 1–7. (https://doi.org/10.1016/j.yhbeh.2015.05.007) org/10.1359/jbmr.2000.15.3.507)
Mohler ML, Bohl CE, Jones A, Coss CC, Narayanan R, He Y, Dong JH, Palmiter RD 2008 Dopamine signaling in the dorsal striatum is essential
Dalton JT & Miller DD 2009 Nonsteroidal Selective Androgen for motivated behaviors: lessons from dopamine-deficient mice.
Receptor Modulators (SARMs): dissociating the anabolic and Annals of the New York Academy of Sciences 1129 35–46. (https://doi.
androgenic activities of the androgen receptor for therapeutic benefit. org/10.1196/annals.1417.003)
Journal of Medicinal Chemistry 52 3597–3617. (https://doi.org/10.1021/ Petroianu A, Veloso DF, Alberti LR, Figueiredo JA & Rodrigues FH
jm900280m) 2010 Is there a relationship between physical performance and
Morimoto M, Amano Y, Oka M, Harada A, Fujita H, Hikichi Y, Tozawa R, orchiectomy? Andrologia 42 302–304. (https://doi.org/10.1111/j.1439-
Yamaoka M & Hara T 2018 Amelioration of sexual behavior and 0272.2009.00997.x)
motor activity deficits in a castrated rodent model with a selective Phoenix CH, Goy R, Gerall A & Young W 1959 Organizing action of
androgen receptor modulator SARM-2f. PLoS ONE 12 e0189480. prenatally administered testosterone propionate on the tissues
(https://doi.org/10.1371/journal.pone.0189480) mediating mating behavior in the female guinea pig. Endocrinology 65
Morin LP & Cummings LA 1981 Effect of surgical or photoperiodic 369–382. (https://doi.org/10.1210/endo-65-3-369)
castration, testosterone replacement or pinealectomy on male Ponnusamy S, Sullivan RD, You D, Zafar N, Yang CH, Thiyagarajan T,
hamster running rhythmicity. Physiology and Behavior 26 825–838. Johnson DL, Barrett ML, Koehler NJ, Star M, et al. 2017 Androgen
(https://doi.org/10.1016/0031-9384(81)90106-2) receptor agonists increase lean mass, improve cardiopulmonary
Musatov S, Chen W, Pfaff DW, Mobbs CV, Yang X-J, Clegg DJ, Kaplitt MG functions and extend survival in preclinical models of Duchenne
& Ogawa S 2007 Silencing of estrogen receptor alpha in the muscular dystrophy. Human Molecular Genetics 26 2526–2540.
ventromedial nucleus of hypothalamus leads to metabolic syndrome. (https://doi.org/10.1093/hmg/ddx150)
PNAS 104 2501–2506. (https://doi.org/10.1073/pnas.0610787104) Pope HG & Katz L 1994 Psychiatric and medical effects of anabolic-
Nilsson ME, Vandenput L, Tivesten Å, Norlén AK, Lagerquist MK, androgenic steroid use: a controlled study of 160 athletes. Archives
Windahl SH, Börjesson AE, Farman HH, Poutanen M, Benrick A, of General Psychiatry 51 375–382. (https://doi.org/10.1001/
et al. 2015 Measurement of a comprehensive sex steroid profile in archpsyc.1994.03950050035004)
rodent serum by high-sensitive gas chromatography-tandem mass Purves-Tyson TD, Handelsman DJ, Double KL, Owens SJ, Bustamante S
spectrometry. Endocrinology 156 2492–2502. (https://doi.org/10.1210/ & Weickert C 2012 Testosterone regulation of sex steroid-
en.2014-1890) related mRNAs and dopamine-related mRNAs in adolescent
Novak CM, Burghardt PR & Levine JA 2012 The use of a running wheel male rat substantia nigra. BMC Neuroscience 13 95. (https://doi.
to measure activity in rodents: relationship to energy balance, org/10.1186/1471-2202-13-95)
general activity, and reward. Neuroscience and Biobehavioral Reviews 36 Purves-Tyson TD, Owens SJ, Double KL, Desai R, Handelsman DJ
1001–1014. (https://doi.org/10.1016/j.neubiorev.2011.12.012) & Weickert CS 2014 Testosterone induces molecular changes in
Nugent BM, Wright CL, Shetty AC, Hodes GE, Lenz KM, Mahurkar A, dopamine signaling pathway molecules in the adolescent male rat
Russo SJ, Devine SE & McCarthy MM 2015 Brain feminization nigrostriatal pathway. PLoS ONE 9 e91151. (https://doi.org/10.1371/
requires active repression of masculinization via DNA methylation. journal.pone.0091151)
Nature Neuroscience 18 690–697. (https://doi.org/10.1038/nn.3988) Purves-Tyson TD, Boerrigter D, Allen K, Zavitsanou K, Karl T, Djunaidi V,
Ogawa S, Chester AE, Hewitt SC, Walker VR, Gustafsson J-Å, Smithies O, Double KL, Desai R, Handelsman DJ & Weickert CS 2015 Testosterone
Korach KS & Pfaff DW 2000 Abolition of male sexual behaviors in attenuates and the selective estrogen receptor modulator, raloxifene,
mice lacking estrogen receptors α and β (αβERKO). PNAS 97 potentiates amphetamine-induced locomotion in male rats. Hormones
14737–14741. (https://doi.org/10.1073/pnas.250473597) and Behavior 70 73–84. (https://doi.org/10.1016/j.yhbeh.2015.02.005)
Ogawa S, Chan J, Gustafsson JÅ, Korach KS & Pfaff DW 2003 Estrogen Quignot N, Arnaud M, Robidel F, Lecomte A, Tournier M, Cren-Olivé C,
increases locomotor activity in mice through estrogen receptor α: Barouki R & Lemazurier E 2012 Characterization of endocrine-
specificity for the type of activity. Endocrinology 144 230–239. (https:// disrupting chemicals based on hormonal balance disruption in male
doi.org/10.1210/en.2002-220519) and female adult rats. Reproductive Toxicology 33 339–352. (https://doi.
Okura T, Koda M, Ando F, Niino N, Ohta S & Shimokata H 2003 org/10.1016/j.reprotox.2012.01.004)
Association of polymorphisms in the estrogen receptor alpha Ramamani A, Aruldhas M & Govindarajulu P 1999 Differential response
gene with body fat distribution. International Journal of Obesity and of rat skeletal muscle glycogen metabolism to testosterone and
Related Metabolic Disorders 27 1020–1027. (https://doi.org/10.1038/ estradiol. Canadian Journal of Physiology and Pharmacology 77 300–304.
sj.ijo.0802378) (https://doi.org/10.1139/y99-016)
Onakomaiya MM, Porter DM, Oberlander JG & Henderson LP 2014 Sex Rana K, Fam BC, Clarke MV, Pang TPS, Zajac JD & MacLean HE 2011
and exercise interact to alter the expression of anabolic androgenic Increased adiposity in DNA binding-dependent androgen receptor
steroid-induced anxiety-like behaviors in the mouse. Hormones and knockout male mice associated with decreased voluntary activity and
Behavior 66 283–297. (https://doi.org/10.1016/j.yhbeh.2014.04.008) not insulin resistance. American Journal of Physiology: Endocrinology

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain
Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access
Journal of F Jardí et al. Androgens and physical activity 238:1 R50
Endocrinology

and Metabolism 301 E767–E778. (https://doi.org/10.1152/ PER2::LUC mice. Scientific Reports 6 e27607. (https://doi.org/10.1038/
ajpendo.00584.2010) srep.27607)
Rana K, Chiu MWS, Russell PK, Skinner JP, Lee NKL, Fam BC, Zajac JD & Sato K, Iemitsu M, Aizawa K & Ajisaka R 2008 Testosterone and DHEA
Maclean HE 2016 Muscle-specific androgen receptor deletion shows activate the glucose metabolism-related signaling pathway in skeletal
limited actions in myoblasts but not in myofibers in different muscles muscle. American Journal of Physiology: Endocrinology and Metabolism
in vivo. Journal of Molecular Endocrinology 57 125–138. (https://doi. 294 E961–E968. (https://doi.org/10.1152/ajpendo.00678.2007)
org/10.1530/JME-15-0320) Sato K, Fujita S, Yamauchi H, Shiroya Y, Kitamura H, Minato K
Raskin K, de Gendt K, Duittoz A, Liere P, Verhoeven G, Tronche F & Motoyuki I 2013 The exercise-induced improvement in
& Mhaouty-Kodja S 2009 Conditional inactivation of androgen hyperglycemia is mediated by DHT produced in the skeletal muscle of
receptor gene in the nervous system: effects on male behavioral and Zucker diabetic fatty rats. Journal of Diabetes and Metabolism 4 239.
neuroendocrine responses. Journal of Neuroscience 29 4461–4470. Sattler F, Bhasin S, He J, Chou CP, Castaneda-Sceppa C, Yarasheski K,
(https://doi.org/10.1523/JNEUROSCI.0296-09.2009) Binder E, Schroeder ET, Kawakubo M, Zhang A, et al. 2011
Rhodes JS, Garland T & Gammie SC 2003 Patterns of brain Testosterone threshold levels and lean tissue mass targets needed to
activity associated with variation in voluntary wheel-running enhance skeletal muscle strength and function: the HORMA trial.
behavior. Behavioral Neuroscience 117 1243–1256. (https://doi. Journals of Gerontology – Series A Biological Sciences and Medical Sciences
org/10.1037/0735-7044.117.6.1243) 66A 122–129. (https://doi.org/10.1093/gerona/glq183)
Rizk A, Robertson J & Raber J 2005 Behavioral performance of tfm mice Schneeberger M, Gomis R & Claret M 2014 Hypothalamic and brainstem
supports the beneficial role of androgen receptors in spatial learning neuronal circuits controlling homeostatic energy balance. Journal of
and memory. Brain Research 1034 132–138. (https://doi.org/10.1016/j. Endocrinology 220 T25–T46. (https://doi.org/10.1530/JOE-13-0398)
brainres.2004.12.002) Schulz KM, Richardson HN, Zehr JL, Osetek AJ, Menard TA &
Roselli CE, Horton LE & Resko JA 1985 Distribution and regulation Sisk CL 2004 Gonadal hormones masculinize and defeminize
of aromatase activity in the rat hypothalamus and limbic system. reproductive behaviors during puberty in the male Syrian hamster.
Endocrinology 117 2471–2477. (https://doi.org/10.1210/endo-117-6- Hormones and Behavior 45 242–249. (https://doi.org/10.1016/j.
2471) yhbeh.2003.12.007)
Roy EJ & Wade GN 1975 Role of estrogens in androgen-induced Schwarz JM, Liang SL, Thompson SM & McCarthy MM 2008 Estradiol
spontaneous activity in male rats. Journal of Comparative and induces hypothalamic dendritic spines by enhancing glutamate
Physiological Psychology 89 573–579. (https://doi.org/10.1037/ release: a mechanism for organizational sex differences. Neuron 58
h0077436) 584–598. (https://doi.org/10.1016/j.neuron.2008.03.008)
Royston SE, Yasui N, Kondilis AG, Lord SV, Katzenellenbogen JA & Sebag IA, Gillis M-A, Calderone A, Kasneci A, Meilleur M, Haddad R,
Mahoney MM 2014 ESR1 and ESR2 differentially regulate daily Noiles W, Patel B & Chalifour LE 2011 Sex hormone control of
and circadian activity rhythms in female mice. Endocrinology 155 left ventricular structure/function: mechanistic insights using
2613–2623. (https://doi.org/10.1210/en.2014-1101) echocardiography, expression, and DNA methylation analyses in
Royston SE, Bunick D & Mahoney MM 2016 Oestradiol exposure early adult mice. American Journal of Physiology: Heart and Circulatory
in life programs daily and circadian activity rhythms in adult mice. Physiology 301 H1706–H1715. (https://doi.org/10.1152/
Journal of Neuroendocrinology 28 1–12. (https://doi.org/10.1111/ ajpheart.00088.2011)
jne.12335) Sharma G, Hu C, Brigman JL, Zhu G, Hathaway HJ & Prossnitz ER
Salahpour A, Ramsey AJ, Medvedev IO, Kile B, Sotnikova TD, 2013 GPER deficiency in male mice results in insulin resistance,
Holmstrand E, Ghisi V, Nicholls PJ, Wong L, Murphy K, et al. 2008 dyslipidemia, and a proinflammatory state. Endocrinology 154
Increased amphetamine-induced hyperactivity and reward in mice 4136–4145. (https://doi.org/10.1210/en.2013-1357)
overexpressing the dopamine transporter. PNAS 105 4405–4410. Silvennoinen M, Rantalainen T & Kainulainen H 2014 Validation of a
(https://doi.org/10.1073/pnas.0707646105) method to measure total spontaneous physical activity of sedentary
Salmen T, Heikkinen A-M, Mahonen A, Kroger H, Komulainen M, and voluntary running mice. Journal of Neuroscience Methods 235
Pallonen H, Saarikoski S, Honkanen R & Maenpaa PH 2003 Relation 51–58. (https://doi.org/10.1016/j.jneumeth.2014.06.027)
of aromatase gene polymorphism and hormone replacement therapy Simonen RL, Perusse L, Rankinen T, Rice T, Rao DC & Bouchard C 2002
to serum estradiol levels, bone mineral density, and fracture risk Familial aggregation of physical activity levels in the Quebec Family
in early postmenopausal women. Annals of Medicine 35 282–288. Study. Medicine and Science in Sports and Exercise 34 1137–1142.
(https://doi.org/10.1080/07853890310006370) (https://doi.org/10.1097/00005768-200207000-00014)
Salmén T, Heikkinen A-M, Mahonen A, Kroger H, Komulainen M, Simonen RL, Rankinen T, Pérusse L, Leon AS, Skinner JS, Wilmore JH,
Saarikoski S, Honkanen R & Maenpaa PH 2000 Early postmenopausal Rao D. & Bouchard C 2003 A dopamine D2 receptor gene
bone loss is associated with PvuII estrogen receptor gene polymorphism and physical activity in two family studies.
polymorphism in Finnish women: effect of hormone replacement Physiology and Behavior 78 751–757. (https://doi.org/10.1016/S0031-
therapy. Journal of Bone and Mineral Research 15 315–321. 9384(03)00084-2)
Sano K, Tsuda MC, Musatov S, Sakamoto T & Ogawa S 2013 Differential Snyder PJ, Peachey H, Hannoush P, Berlin JA, Loh L, Lenrow DA,
effects of site-specific knockdown of estrogen receptor α in the Holmes JH, Dlewati A, Santanna J, Rosen CJ, et al. 1999 Effect of
medial amygdala, medial pre-optic area, and ventromedial nucleus testosterone treatment on body composition and muscle strength
of the hypothalamus on sexual and aggressive behavior of male in men over 65 years of age. Journal of Clinical Endocrinology and
mice. European Journal of Neuroscience 37 1308–1319. (https://doi. Metabolism 84 2647–2653. (https://doi.org/10.1210/jcem.84.8.5885)
org/10.1111/ejn.12131) Snyder PJ, Bhasin S, Cunningham GR, Matsumoto AM, Stephens-
Sano K, Nakata M, Musatov S, Morishita M, Sakamoto T, Tsukahara S Shields AJ, Cauley JA, Gill TM, Barrett-Connor E, Swerdloff RS,
& Ogawa S 2016 Pubertal activation of estrogen receptor α in the Wang C, et al. 2016 Effects of testosterone treatment in older
medial amygdala is essential for the full expression of male social men. New England Journal of Medicine 374 611–624. (https://doi.
behavior in mice. PNAS 113 7632–7637. (https://doi.org/10.1073/ org/10.1056/NEJMoa1506119)
pnas.1524907113) Srinivas-Shankar U, Roberts SA, Connolly MJ, O’Connell MDL, Adams JE,
Sasaki H, Hattori Y, Ikeda Y, Kamagata M, Iwami S, Yasuda S, Tahara Y Oldham JA & Wu FCW 2010 Effects of testosterone on muscle
& Shibata S 2016 Forced rather than voluntary excercise entrains strength, physical function, body composition, and quality of life
peripheral clocks via a corticosterone/noradrenaline increase in in intermediate-frail and frail elderly men: a randomized, double-

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access
Journal of F Jardí et al. Androgens and physical activity 238:1 R51
Endocrinology

blind, placebo-controlled study. Journal of Clinical Endocrinology and Uchida M, Palmateer JM, Herson PS, DeVries AC, Cheng J & Hurn PD
Metabolism 95 639–650. (https://doi.org/10.1210/jc.2009-1251) 2009 Dose-dependent effects of androgens on outcome after focal
Stanić D, Dubois S, Chua HK, Tonge B, Rinehart N, Horne MK & cerebral ischemia in adult male mice. Journal of Cerebral Blood
Boon WC 2014 Characterization of aromatase expression in the Flow and Metabolism 29 1454–1462. (https://doi.org/10.1038/
adult male and female mouse brain. I. Coexistence with oestrogen jcbfm.2009.60)
receptors α and β, and androgen receptors. PLoS ONE 9 e90451. Unger EK, Burke KJ, Yang CF, Bender KJ, Fuller PM & Shah NM 2015
(https://doi.org/10.1371/journal.pone.0090451) Medial amygdalar aromatase neurons regulate aggression in
Stefan N, Vozarova B, Del Parigi A, Ossowski V, Thompson D, Hanson R, both sexes. Cell Reports 10 453–463. (https://doi.org/10.1016/j.
Ravussin E & Tataranni P 2002 The Gln223Arg polymorphism of the celrep.2014.12.040)
leptin receptor in Pima Indians: influence on energy expenditure, Vanderschueren D, Laurent MR, Claessens F, Gielen E, Lagerquist MK,
physical activity and lipid metabolism. International Journal of Obesity Vandenput L, Börjesson AE & Ohlsson C 2014 Sex steroid actions in
26 1629–1632. (https://doi.org/10.1038=sj.ijo.0802161) male bone. Endocrine Reviews 35 906–960. (https://doi.org/10.1210/
Stern JS & Johnson PR 1977 Spontaneous activity and adipose cellularity er.2014-1024)
in the genetically obese Zucker rat (fafa). Metabolism 26 371–380. Vanzyl CG, Noakes TD & Lambert MI 1995 Anabolic-androgenic steroid
(https://doi.org/10.1016/0026-0495(77)90104-4) increases running endurance in rats. Medicine and Science in Sports and
Stern JJ & Murphy M 1971 The effects of cyproterone acetate on the Exercise 27 1385–1389.
spontaneous activity and seminal vesicle weight of male rats. Walsh S, Zmuda JM, Cauley JA, Shea PR, Metter EJ, Hurley BF, Ferrell RE
Journal of Endocrinology 50 441–443. (https://doi.org/10.1677/ & Roth SM 2005 Androgen receptor CAG repeat polymorphism is
joe.0.0500441) associated with fat-free mass in men. Journal of Applied Physiology 98
Storer TW, Basaria S, Traustadottir T, Harman SM, Pencina K, Li Z, 132–137. (https://doi.org/10.1152/japplphysiol.00537.2004)
Travison TG, Miciek R, Tsitouras P, Hally K, et al. 2016 Effects of Wang L, Kang Y, Zhang G, Zhang Y, Cui R, Yan W, Tan H, Li S, Wu B,
testosterone supplementation for 3-years on muscle performance and Cui H, et al. 2016 Deficits in coordinated motor behavior and
physical function in older men. Journal of Clinical Endocrinology and in nigrostriatal dopaminergic system ameliorated and VMAT2
Metabolism 102 jc20162771. (https://doi.org/10.1210/jc.2016-2771) expression up-regulated in aged male rats by administration of
Strath SJ, Kaminsky LA, Ainsworth BE, Ekelund U, Freedson PS, Gary RA, testosterone propionate. Experimental Gerontology 78 1–11. (https://
Richardson CR, Smith DT & Swartz AM 2013 Guide to the assessment doi.org/10.1016/j.exger.2016.03.003)
of physical activity: clinical and research applications: a scientific Watai K, Tsuda M, Nakata M, Toda K & Ogawa S 2007 Analyses of
statement from the American Heart Association. Circulation 128 running wheel activity (RWA) in aromatase-knockout (ArKO)
2259–2279. (https://doi.org/10.1161/01.cir.0000435708.67487.da) mice. Neuroscience Research 58S S108. (https://doi.org/10.1016/j.
Stubbe JH, Boomsma DI, Vink JM, Cornes BK, Martin NG, Skytthe A, neures.2007.06.1200)
Kyvik KO, Rose RJ, Kujala UM, Kaprio J, et al. 2006 Genetic Wen CP, Wai JPM, Tsai MK, Yang YC, Cheng TYD, Lee MC, Chan HT,
influences on exercise participation in 37.051 twin pairs from Tsao CK, Tsai SP & Wu X 2011 Minimum amount of physical activity
seven countries. PLoS ONE 1 e22. (https://doi.org/10.1371/journal. for reduced mortality and extended life expectancy: a prospective
pone.0000022) cohort study. Lancet 378 1244–1253. (https://doi.org/10.1016/S0140-
Tajar A, Huhtaniemi IT, O’Neill TW, Finn JD, Pye SR, Lee DM, Bartfai G, 6736(11)60749-6)
Boonen S, Casanueva FFF, Forti G, et al. 2012 Characteristics of White JP, Gao S, Puppa MJ, Sato S, Welle SL & Carson JA 2013
androgen deficiency in Late-onset hypogonadism: results from the Testosterone regulation of Akt/mTORC1/FoxO3a signaling in skeletal
European Male Aging Study (EMAS). Journal of Clinical Endocrinology muscle. Molecular and Cellular Endocrinology 365 174–186. (https://
and Metabolism 97 1508–1516. (https://doi.org/10.1210/jc.2011- doi.org/10.1016/j.mce.2012.10.019)
2513) WHO 2009 Global health risks: mortality and burden of disease
Takahashi LK 2014 Olfactory systems and neural circuits that modulate attributable to selected major risks. Bulletin of the World Health
predator odor fear. Frontiers in Behavioral Neuroscience 8 72. (https:// Organization 87 646–646. (https://doi.org/10.2471/BLT.09.070565)
doi.org/10.3389/fnbeh.2014.00072) WHO 2010 Global Recommendations on Physical Activity for Health, p
Takeshita H, Yamamoto K, Nozato S, Inagaki T, Tsuchimochi H, 60. Geneva, Switzerland: World Health Organization. (https://doi.
Shirai M, Yamamoto R, Imaizumi Y, Hongyo K, Yokoyama S, et al. org/10.1080/11026480410034349)
2017 Modified forelimb grip strength test detects aging-associated Windahl SH, Andersson N, Börjesson AE, Swanson C, Svensson J,
physiological decline in skeletal muscle function in male mice. Movérare-Skrtic S, Sjögren K, Shao R, Lagerquist MK, et al. 2011
Scientific Reports 7 42323. (https://doi.org/10.1038/srep42323) Reduced bone mass and muscle strength in male 5α-reductase type
Tanehkar F, Rashidy-Pour A, Vafaei AA, Sameni HR, Haghighi S, Miladi- 1 inactivated mice. PLoS ONE 6 e21402. (https://doi.org/10.1371/
Gorji H, Motamedi F, Akhavan MM & Bavarsad K 2013 Voluntary journal.pone.0021402)
exercise does not ameliorate spatial learning and memory deficits Wise RA 2004 Dopamine, learning and motivation. Nature Reviews
induced by chronic administration of nandrolone decanoate in Neuroscience 5 483–494. (https://doi.org/10.1038/nrn1406)
rats. Hormones and Behavior 63 158–165. (https://doi.org/10.1016/j. Wollnik F & Turek FW 1988 Estrous correlated modulations of circadian
yhbeh.2012.10.003) and ultradian wheel-running activity rhythms in LEW/Ztm rats.
Teske JA, Perez-Leighton CE, Billington CJ & Kotz CM 2014 Physiology and Behavior 43 389–396. (https://doi.org/10.1016/0031-
Methodological considerations for measuring spontaneous physical 9384(88)90204-1)
activity in rodents. American Journal of Physiology: Regulatory, Wu FCW, Tajar A, Pye SR, Silman AJ, Finn JD, O’Neill TW, Bartfai G,
Integrative and Comparative Physiology 306 R714–R721. (https://doi. Casanueva F, Forti G, Giwercman A, et al. 2008 Hypothalamic-
org/10.1152/ajpregu.00479.2013) pituitary-testicular axis disruptions in older men are differentially
Townsend EA, Miller VM & Prakash YS 2012 Sex differences and sex linked to age and modifiable risk factors: the European male aging
steroids in lung health and disease. Endocrine Reviews 33 1–47. study. Journal of Clinical Endocrinology and Metabolism 93 2737–2745.
(https://doi.org/10.1210/er.2010-0031) (https://doi.org/10.1210/jc.2007-1972)
Troiano RP, Berrigan D, Dodd KW, Mâsse LC, Tilert T & Mcdowell M 2008 Wu MV, Manoli DS, Fraser EJ, Coats JK, Tollkuhn J, Honda SI, Harada N
Physical activity in the United States measured by accelerometer. & Shah NM 2009 Estrogen masculinizes neural pathways and
Medicine and Science in Sports and Exercise 40 181–188. (https://doi. sex-specific behaviors. Cell 139 61–72. (https://doi.org/10.1016/j.
org/10.1249/mss.0b013e31815a51b3) cell.2009.07.036)

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain
Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access
Journal of F Jardí et al. Androgens and physical activity 238:1 R52
Endocrinology

Wu J, Movérare-Skrtic S, Börjesson AE, Lagerquist MK, Sjögren K, regulates body weight. Journal of Clinical Investigation 125 2861–2876.
Windahl SH, Koskela A, Grahnemo L, Islander U, Wilhelmson AS, (https://doi.org/10.1172/JCI80941)
et al. 2016 Enzalutamide reduces the bone mass in the axial but not Xu Y, O’Malley BW & Elmquist JK 2017 Brain nuclear receptors and body
the appendicular skeleton in male mice. Endocrinology 157 969–977. weight regulation. Journal of Clinical Investigation 127 1–9. (https://
(https://doi.org/10.1210/en.2015-1566) doi.org/10.1172/JCI88891)
Xirouchaki CE, Mangiafico SP, Bate K, Ruan Z, Huang AM, Yoo YE & Ko CP 2012 Dihydrotestosterone ameliorates degeneration
Tedjosiswoyo BW, Lamont B, Pong W, Favaloro J, Blair AR, et al. in muscle, axons and motoneurons and improves motor function
2016 Impaired glucose metabolism and exercise capacity with in amyotrophic lateral sclerosis model mice. PLoS ONE 7 e37258.
muscle-specific glycogen synthase 1 (gys1) deletion in adult (https://doi.org/10.1371/journal.pone.0037258)
mice. Molecular Metabolism 5 221–232. (https://doi.org/10.1016/j. Yu IC, Lin HY, Liu NC, Sparks JD, Yeh S, Fang LY, Chen L & Chang C
molmet.2016.01.004) 2013 Neuronal androgen receptor regulates insulin sensitivity via
Xu Y, Nedungadi TP, Zhu L, Sobhani N, Irani BG, Davis KE, Zhang X, suppression of hypothalamic NF-KB-mediated PTP1B expression.
Zou F, Gent LM, Hahner LD, et al. 2011 Distinct hypothalamic Diabetes 62 411–423. (https://doi.org/10.2337/db12-0135)
neurons mediate estrogenic effects on energy homeostasis and Zhao C, Fujinaga R, Tanaka M, Yanai A, Nakahama KI & Shinoda K 2007
reproduction. Cell Metabolism 14 453–465. (https://doi.org/10.1016/j. Region-specific expression and sex-steroidal regulation on aromatase
cmet.2011.08.009) and its mRNA in the male rat brain: immunohistochemical and in
Xu P, Cao X, He Y, Zhu L, Yang Y, Saito K, Wang C, Yan X, Hinton AO, situ hybridization analyses. Journal of Comparative Neurology 500
Zou F, et al. 2015 Estrogen receptor-α in medial amygdala neurons 557–573. (https://doi.org/10.1002/cne.21193)

Received in final form 29 March 2018


Accepted 9 May 2018
Accepted Preprint published online 9 May 2018

http://joe.endocrinology-journals.org © 2018 Society for Endocrinology


https://doi.org/10.1530/JOE-18-0125 Published by Bioscientifica Ltd.
Printed in Great Britain Downloaded from Bioscientifica.com at 01/28/2020 01:32:50PM
via free access

You might also like