ArticlePDF AvailableLiterature Review

Relationship between Testosterone and Sarcopenia in Older-Adult Men: A Narrative Review

MDPI
Journal of Clinical Medicine
Authors:

Abstract and Figures

Age-related decline in testosterone is known to be associated with various clinical symptoms among older men and it is possible that the accompanying decline in muscle mass and strength might lead to a decline in motor and physical functions. Sarcopenia is an important pathophysiological factor associated with frailty in older adults and is diagnosed in older adults as a decrease in muscle strength, muscle mass, and walking speed, which can lead to a significant decline in the quality of life and shortened healthy life expectancy. Testosterone directly interacts with the androgen receptor expressed in myonuclei and satellite cells and is also indirectly associated with muscle metabolism through various cytokines and molecules. Currently, significant correlations between testosterone and frailty in men have been confirmed by numerous cross-sectional studies. Many randomized control studies have also supported the beneficial effect of testosterone replacement therapy (TRT) on muscle volume and strength among men with low to normal testosterone levels. In the world’s aging society, TRT can be a tool for preventing the onset of sarcopenia in older-adult men. This narrative review aims to show the relationship between the decline in testosterone with age, sarcopenia, and frailty, as well as the effects of testosterone replacement therapy on muscle mass and strength.
Content may be subject to copyright.
J. Clin. Med. 2022, 11, 6202. https://doi.org/10.3390/jcm11206202 www.mdpi.com/journal/jcm
Review
Relationship between Testosterone and Sarcopenia
in Older-Adult Men: A Narrative Review
Kazuyoshi Shigehara *, Yuki Kato, Kouji Izumi and Atsushi Mizokami
Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science,
13-1, Takaramachi, Kanazawa 920-8641, Japan
* Correspondence: kshigehara0415@yahoo.co.jp; Tel.: +81-76-265-2393; Fax: +81-76-234-4263
Abstract: Age-related decline in testosterone is known to be associated with various clinical symp-
toms among older men and it is possible that the accompanying decline in muscle mass and strength
might lead to a decline in motor and physical functions. Sarcopenia is an important pathophysio-
logical factor associated with frailty in older adults and is diagnosed in older adults as a decrease in
muscle strength, muscle mass, and walking speed, which can lead to a significant decline in the
quality of life and shortened healthy life expectancy. Testosterone directly interacts with the andro-
gen receptor expressed in myonuclei and satellite cells and is also indirectly associated with muscle
metabolism through various cytokines and molecules. Currently, significant correlations between
testosterone and frailty in men have been confirmed by numerous cross-sectional studies. Many
randomized control studies have also supported the beneficial effect of testosterone replacement
therapy (TRT) on muscle volume and strength among men with low to normal testosterone levels.
In the world’s aging society, TRT can be a tool for preventing the onset of sarcopenia in older-adult
men. This narrative review aims to show the relationship between the decline in testosterone with
age, sarcopenia, and frailty, as well as the effects of testosterone replacement therapy on muscle
mass and strength.
Keywords: testosterone; sarcopenia; frailty; muscle
1. Introduction
The aging of the population has been become a worldwide problem and maintaining
and even improving the quality of life (QOL) of middle-aged and older-adult men has
become an important issue. As the population ages, healthy life expectancy becomes in-
creasingly more important than mean life expectancy. Life expectancy refers to the period
from birth to death and includes periods requiring long-term care, while healthy life expec-
tancy indicates the period without significant health issues in daily life. In many developed
countries, the difference between the mean life expectancy and the healthy life expectancy
tends to be greater and extending healthy life expectancy has become a clinical concern.
In men, serum testosterone levels decrease with age by 2–3% annually, a decline as-
sociated with specific symptoms of late-onset hypogonadism (LOH) syndrome [1], whose
various clinical signs and symptoms include decreased libido and sexual desire, muscle
weakness, increased visceral fat, obesity, osteoporosis, deterioration of insulin resistance,
and dyslipidemia, which are significantly associated with aging [1–4]. These clinical signs
and symptoms can often impair the QOL of middle-aged and older-adult men and are
becoming a serious issue in the present aging society. Testosterone replacement therapy
(TRT) is a widely accepted tool for improving these clinical conditions in hypogonadal
men and its clinical use has increased substantially over the past several years [1].
Citation: Shigehara, K.; Kato, Y.;
Izumi, K.; Mizokami, A.
Relationship between Testosterone
and Sarcopenia in Older-Adult Men:
A Narrative Review. J. Clin. Med.
2022, 11, 6202. https://doi.org/
10.3390/jcm11206202
Academic Editor: Wing Hoi Cheung
Received: 30 September 2022
Accepted: 18 October 2022
Published: 20 October 2022
Publisher’s Note: MDPI stays neu-
tral with regard to jurisdictional
claims in published maps and institu-
tional affiliations.
Copyright: © 2022 by the authors. Li-
censee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and con-
ditions of the Creative Commons At-
tribution (CC BY) license (https://cre-
ativecommons.org/licenses/by/4.0/).
J. Clin. Med. 2022, 11, 6202 2 of 13
Sarcopenia is an important pathophysiological factor of frailty in older adults and is
diagnosed with a decrease in muscle strength, muscle mass, and low physical perfor-
mance [5]. Sarcopenia and frailty are significantly associated with an increased risk of falls
and fractures in older-adult men, which can lead to a serious decrease in QOL and short-
ening of healthy life expectancy [6,7]. Preventive measures are, therefore, required.
Testosterone is an important hormone for maintaining skeletal muscle mass and
strength in men and numerous previous studies have suggested that testosterone defi-
ciency is significantly associated with the onset of sarcopenia. The present review sum-
marizes the current evidence on the relationship between testosterone and sarcope-
nia/frailty. The review also investigated whether TRT for hypogonadal men with sarco-
penia can improve muscle mass, strength, and physical function.
2. Materials and Methods
A review of the PubMed, MEDLINE, and EMBASE databases was conducted to
search for original articles, systematic reviews, and meta-analyses under following key-
words: “testosterone” or “hypogonadism” or “sarcopenia” or “frailty” or “muscle.” There
were no limitations in terms of language, publication status, or study design. Papers pub-
lished between January 1990 and October 2020 were collected. We also checked the refer-
ences of systematic reviews and meta-analyses carefully to identify additional original
articles for inclusion. Two reviewers screened the search results and the data were col-
lected in June 2022.
The papers suitable for the topic “testosterone and sarcopenia” from the journal da-
tabases were chosen for the present analysis. For “efficacy of testosterone replacement
therapy in sarcopenia”, we reviewed papers published since 2010.
3. Sarcopenia: Definition and Etiology
One of the phenomena of human aging is the progressive decline in skeletal muscle
mass, which can result in negative effects on physical fitness and function. The prevalence
of sarcopenia is ~5–50% in older adults aged 65 years and older [8–10]. In 1989, Irwin Ros-
enberg proposed the term “sarcopenia” (from the Greek “sarx” for flesh and “penia” for
deficiency) for age-related loss of muscle mass [11]. Currently, sarcopenia is defined as
age-related loss of skeletal muscle mass and strength. Various differing definitions of sar-
copenia have been proposed; however, there is still no widely accepted definition [12].
Baumgartner et al. first defined sarcopenia as a decline of less than 2 standard deviations
below the mean of a young reference group in appendicular skeletal muscle mass [13].
Since then, the algorithm proposed by the European Working Group on Sarcopenia in
Older People (the presence of both low muscle mass and low muscle strength or perfor-
mance) [14] and by the Foundation for the National Institutes of Health sarcopenia project
(appendicular lean mass adjusting for body mass index to define low muscle mass) [15]
have been generally used for diagnosing sarcopenia. Recently, the Asian working group
for sarcopenia suggested an alternative algorithm (Figure 1) [16]. This Asian consensus
has plenty in common with the European consensus and the cases with decreased walking
speed or grip strength are defined as individuals with decreased muscle performance.
Those cases accompanied by decreased muscle mass are defined as having sarcopenia.
According to the Asian consensus, the reported prevalence for sarcopenia is 16.5% for
men and 19.9% for women in Japan [17], which is likely to be approximately similar to
that reported previously in other countries [18–20].
J. Clin. Med. 2022, 11, 6202 3 of 13
Figure 1. Algorithm for sarcopenia diagnosis (the Asian working group for sarcopenia).
For certain cases, the cause of the sarcopenia can be clearly identified, whereas no
clear cause can be determined in other cases. Sarcopenia caused only by aging is classified
as “primary” (age related) and sarcopenia caused by activities of daily living, nutrition,
and illness is classified as “secondary sarcopenia” (Table 1). The most common cause of
muscle weakness in older adults is age-related muscle atrophy. In general, skeletal muscle
decreases by 25–30% and muscle strength decreases by 30–40% in individuals in their 70s
when compared with those in their 20s, with muscle mass decreasing by approximately
1–2% every year after 50 years of age [21]. In addition, older-adult men have an increased
risk of developing sarcopenia through various factors, such as lifestyle changes, less exer-
cise, more physical illnesses (severe organ failure, neuromuscular disease, inflammatory
disease, malignant tumors, etc.), undernourishment, and appetite loss. Therefore, the eti-
ology of sarcopenia is often assumed to be multifactorial [22]. Sarcopenia develops con-
currently with changes in hormones (testosterone and growth hormones) and inflamma-
tory cytokines involved in the muscle metabolism due to these causes. In particular, tes-
tosterone is significantly correlated with maintaining bone strength, muscle mass, and
muscle strength among men and it has been found that the pathogenesis of sarcopenia in
men might be associated with testosterone decline with aging.
Table 1. Classification of sarcopenia by causes.
Primary Sarcopenia
Age-related No clear causes other than aging
Secondary Sarcopenia
Daily living-related Bedridden, lack of exercise, ataxia, weight loss
Nutrition-related Malabsorption, gastrointestinal disease, appetite loss, lack
of energy, low protein intake
Disease-related Severe
organ failure, inflammatory diseases, malignancies,
endocrine diseases
4. Testosterone and Sarcopenia
4.1. Testosterone and Muscle Metabolism
More than 95% of serum testosterone is produced by the Leydig cells of the testes
through stimulation by LH from the pituitary gland in males [23]. Testosterone is mostly
bound to sex-hormone-binding globulin or albumin and 1–2% exists in free form; how-
ever, it binds loosely to albumin and can easily become free form [24]. Free testosterone
(FT) is taken up into target cells through the cell membrane and binds to androgen recep-
tor (AR) in the cytoplasm. Testosterone bound to AR is converted to dihydrotestosterone
J. Clin. Med. 2022, 11, 6202 4 of 13
by 5α-reductase. Testosterone and DHT bind to the same AR to form a dimeric complex
and this dimer binds to specific sites on DNA and activates target genes, resulting in the
expression of androgenic actions [25]. In general, the length of the CAG repeats present in
the AR gene shows an inverse relationship, with AR susceptibility and length of their re-
peats differing between the races, with the length increasing in the order of black people,
white people, and Asian people [26].
There are numerous ARs in muscle tissue and testosterone plays an important role
in maintaining muscle mass and strength. It is, therefore, logical to assume that age-re-
lated testosterone decreases are closely associated with the onset of sarcopenia in men.
Conversely, the anabolic effects of testosterone on muscle hypertrophy have been well
established [27].
In human studies, testosterone directly interacts with AR expressed in myonuclei and
satellite cells [28,29], which is a major source for the establishment of hypertrophying
muscle fibers (Figure 2). Testosterone has a potential effect on myogenesis and muscle
hypertrophy by increasing protein synthesis and inhibiting protein degradation in muscle
cells [30,31] and then promoting mitotic activity and differentiation of satellite cells
[29,32]. Numerous in vitro studies have demonstrated the anabolic actions of testosterone
through increases in insulin-like growth factor-1 expression [33,34], beta-catenin and T-
cell factor-4 pathway signaling [35], regulation of peroxisome proliferator-activated re-
ceptor-gamma coactivator 1 alpha, and p38 mitogen-activated protein kinases [36] and
stimulating the hypertrophy of L6 myoblasts in a signal cascade dependent on Ark and
mammalian target of rapamycin [37]. The mechanism by which low testosterone levels
cause muscle atrophy is also being clarified. The catabolic action of testosterone has been
described through the enhancement of muscle atrophy-F-box (atrogin-1) and muscle
RING-finger protein-1 expression [38]. Moreover, an increase in hypertrophied visceral
fat due to testosterone decline contributes to an increase in certain inflammatory cyto-
kines, such as interleukin-6 and tumor necrosis factor (TNF-α), which have catabolic ef-
fects on skeletal muscle [39].
Figure 2. Molecular mechanisms in the development of sarcopenia. IGF-1, insulin-like growth fac-
tor–1; mTOR, mammalian target of rapamycin; TCF-4, T-cell factor-4; PGC-1α, peroxisome prolifer-
ator-activated receptor-gamma coactive tor 1 alpha; MAPK, p38 mitogen-activated protein kinases;
TNF-α, tumor necrosis factor; IL-6, interleukin-6; MuRF-1, muscle atrophy-F-box (atrogin-1) and
muscle RING-finger protein-1. An upward red arrow indicates “increase”, whereas a downward
one indicates "decrease".
J. Clin. Med. 2022, 11, 6202 5 of 13
4.2. Clinical Effects of Testosterone for Muscle
Androgen deprivation therapy (ADT) for prostate cancer can result in decreased
muscle mass and muscle weakness. A prospective study that included 79 patients with
prostate cancer and employed a 12-month ADT reduced the participants’ lean body mass
by 3.8% and increased their body fat percentage by 11% [40]. According to a report exam-
ining 39 patients with prostate cancer, the muscle mass of the rectus femoris, sartorius,
and quadriceps estimated using computed tomography after ADT for 14–20 weeks was
21.8% and 15.4%, and 16.6%, respectively [41]. In general, patients with prostate cancer
who undergo ADT are reported to have 3.0–6.0% lower muscle mass and 15–17% lower
muscle strength than healthy individuals of the same age [42]. These findings suggest that
testosterone decline with age is a trigger for muscle loss among older-adult men.
Several studies have demonstrated an association between serum testosterone levels
and muscle mass and strength in men [43–49]. Appendicular muscle mass was signifi-
cantly correlated with serum FT levels in non-Hispanic white men in a cross-sectional
study from the New Mexico Aging Process Study [43]. In 403 men from The Netherlands,
bioavailable testosterone and luteinizing hormone had a significant correlation with grip
and leg extensor strength [44]. The MINOS cohort study that included 845 French men
also found that the group with the lowest appendicular muscle mass had a significantly
lower FT level [45]. A previous National Health and Nutrition Examination Survey study
of men revealed that higher testosterone levels at physiologic levels were associated with
higher body lean mass and lower body fat mass [46]. A cross-sectional study that included
922 men aged over 60 years found that weaker muscle strength was observed in the men
within the lowest tertile of FT compared with those in the highest tertile (adjusted odds
ratio: 2.28; 95% CI 1.33–3.91) [47]. A recent study that investigated the association between
serum testosterone levels and body composition among 3875 men in China found a posi-
tive correlation between testosterone levels and appendicular lean mass index [48]. A re-
cent systematic review also reported that testosterone could have a potential effect on
muscle mass and strength [49]. Although there are a few studies that failed to identify an
effect for testosterone on muscle strength [50,51], current evidence has likely established
a positive correlation between testosterone and muscle condition.
4.3. Evidence of Testosterone Decrease in Frailty/Sarcopenia
Numerous cross-sectional studies have confirmed significant correlations between
testosterone and frailty in men [52–54]. A Massachusetts cohort study that included 646
men aged 50–86 years investigated the relationship between testosterone and frailty and
its components [52]. Although no association was observed between total testosterone
(TT) or FT levels and the frailty phenotype, there was a significant association between TT
levels and the frailty components of grip strength and physical activity. Cross-sectional
data from the Toledo Study for Healthy Aging that included 552 men showed that the risk
of frailty decreases linearly with testosterone levels (adjusted OR 2.9 (95% CI 1.6–5.1) and
1.6 (95% CI 1.0–2.5) in TT and FT, respectively) [53]. Another cross-sectional study based
on data from the Longitudinal Aging Study Amsterdam (LASA) that included 623 men
also suggested a potential correlation between low TT or bioavailable testosterone levels
and impaired mobility and low muscle strength in men [54]. In a study of 461 individuals
aged 60 years and older, a low FT value (<243 pmol/L) was a significant risk factor for
developing frailty [55]. A recent meta-analysis that included 11 studies reported that TT
(OR 1.37, 95% CI 1.09–1.72) and FT (OR 1.55, 1.06–2.25) were significantly associated with
frailty in older men [56].
A number of longitudinal studies have found an equivocal future risk for developing
frailty and sarcopenia due to low baseline testosterone [57–59]. A longitudinal study that
included 957 community-dwelling adult men in Japan demonstrated that low calculated
FT (OR 2.14, 95% CI 1.06–4.33) and FT (OR 1.83, 95% CI 1.04–3.22) were associated with
the onset of sarcopenia [57]. Another report that included 1445 men from the Framingham
J. Clin. Med. 2022, 11, 6202 6 of 13
Offspring Study revealed that low FT levels were significantly associated with the inci-
dence of mobility, limiting its progression, but was not associated with subjective health,
usual walking speed, or handgrip strength after 6.6 years of follow-up [58]. A longitudinal
study that included 486 men from LASA and 1071 well-functioning men from the Health,
Aging and Body Composition study demonstrated that baseline FT was not associated
with changes in physical performance, walking speed, or muscle strength after 3 years of
follow-up [59]. Further studies are needed to conclude whether low testosterone levels
predict the progression and development of incident frailty and sarcopenia.
5. Efficacy of Testosterone Replacement Therapy for Sarcopenia
5.1. Indication of Testosterone Replacement Therapy
The indication of TRT requires the presence of low serum testosterone level. How-
ever, the cut-off value of serum testosterone for a diagnosis of hypogonadism is still con-
troversial, with multiple international societies’ recommendations [60]. The diagnosis of
hypogonadism for the recommendation of TRT in the guidelines of the Consensus Com-
mittee of the American Urological Association (AUA) is TT ≤  3.0 ng/mL [61]. On the other
hand, according to the International Society for Sexual Medicine (ISSM), the International
Society for the Study of Aging Males (ISSAM), and the European Association of Urology
(EAU), serum TT levels above 12 nmol/L (346 ng/dL) are normal and TT levels below 8
nmol/L (231 ng/dL) indicate hypogonadism, meaning that TRT may be appropriate
[3,4,62–64]. In cases with borderline TT values of 8–12 nmol/L, hypogonadism should be
diagnosed with calculated FT values.
5.2. Efficacy of Testosterone Replacement Therapy for Sarcopenia
The randomized controlled trials (RCTs) published since 2010 are summarized in Ta-
ble 2 [50,65–82]. Their results varied by target population, type of testosterone formula-
tion, and testosterone dosage. Many of the RCTs investigated the effects on muscle of TRT
among men with low to normal testosterone levels and 15 of 19 RCTs supported certain
merits of TRT on muscle volume [50,67–70,72–77,79–82]. The other four studies, however,
failed to demonstrate that TRT contributes to improving muscle mass or strength
[65,66,71,78]. In one of the RCTs, the study population consists of patients with opioid-
induced hypogonadism, which was a specific population differing from the LOH syn-
drome [65]. The other three studies investigated the efficacy of exercise and/or diet added
to TRT on muscle mass and strength but did not study the direct effects of TRT in isolation
[66,72,78]. These findings suggest that monolithic TRT for hypogonadal men can contrib-
ute to improving muscle mass and strength. However, certain clinical interventions, such
as exercise and diet added to TRT, are likely to be the most important factor for maintain-
ing muscle function among older-adult men.
Table 2. Randomized control trials to investigate the effects of testosterone replacement therapy
(TRT) on muscle (published since 2010).
Author Year Subjects Number TRT Regimens
(Add-on Therapy) Effects Ref.
Kolind
(Denmark) 2022
Hypogonadal men
with opioid-treated
chronic pain
(TT < 12 nmol/L)
41 TRT 20
placebo 21
TU 1000 mg,
intramuscular
for 24 weeks
TRT did not improve muscle function
(leg-press maximal voluntary
contraction, leg
extension power and
handgrip strength).
[65]
Barnouin
(USA) 2021
Hypogonadal men
with obesity
(TT < 10.4 nmol/L)
83 TRT 42
placebo 41
T gel daily
for 6 months
(diet + exercise)
TRT might attenuate the weight loss–
induced reduction in muscle mass.
There was no significant difference in
muscle strength between the two
groups.
[66]
J. Clin. Med. 2022, 11, 6202 7 of 13
Chasland
(Australia) 2021
Men with obesity and
low-
normal serum TT
(TT 6–14 nmol/L)
80 TRT 40
placebo 40
T gel 100 mg/day
for 23 weeks
(exercise)
TRT
increased total, leg, and arm lean
mass but did not affect aerobic
capacity (Vo2peak) and muscle
strength.
[67]
Glintborg
(Denmark) 2020
Men with opioid-
induced
hypogonadism
(TT < 12 nmol/L)
41 TRT 20
placebo 21
TU 1000 mg,
intramuscular
for 24 weeks
TRT increased lean body mass. [68]
Gagliano-Juca
(USA) 2018
Older men with
mobility limitations
(TT <
350 ng/dL or FT
< 50 pg/mL)
99 TRT 46
placebo 53
T gel 100 mg/day
for 6 months
TRT improved muscle strength and
physical function (assessed by loaded
stair-climbing power).
[69]
Storer
(USA) 2017
Eugonadal and
hypogonadal men
(TT 100–
400 ng/dL or
FT < 50 pg/mL)
256 TRT 135
placebo 121
T gel 75 mg/day
for 3 years
TRT strengthened chest-
press strength
and power, and leg-press power. [70]
Ng Tang Fui
(Australia) 2016
Hypogonadal men
with obesity
(TT < 12 nmol/L)
100 TRT 49
placebo 51
TU 1000 mg
intramuscular
for 56 weeks (diet)
TRT did not increase muscle volume
but did
attenuate the reduction in lean
mass by diet compared with the
controls.
[71]
Dias
(USA) 2016 Hypogonadal men
(TT < 350 ng/dL) 39
TRT 13
placebo 9
other 13
T gel 50 mg/day
for 12 months
TRT improved knee strength and fast
gait at 12 months compared with
baseline.
[72]
Konaka
(Japan) 2016 Hypogonadal men
(FT < 10.8 pg/mL) 334 TRT 169
control 165
TE 250 mg/4 weeks
for 52 weeks
TRT improved muscle volume and
grip power. [73]
Magnussen
(Denmark) 2016
Hypogonadal men
with DM
(BioT < 7.3 nmol/L)
43 TRT 22
placebo 21
T gel 50 mg/day
for 24 weeks TRT increased lean body mass. [74]
Sinclair
(Australia) 2016
Hypogonadal men
with cirrhosis
(TT <
12 nmol/L or FT
< 230 pmol/L)
101 TRT 50
placebo 51
TU 1000 mg/6–12
weeks intramuscular
for 12 months
TRT increased total lean body and
appendicular lean muscle mass. [75]
Borst
(USA) 2014 Hypogonadal men
(TT ≤ 300 ng/dL) 60 TRT 31
placebo 29
TE 125 mg/weeks
I intramuscular for 12
months
(finasteride)
TRT
increased upper and lower body
muscle strength by 8–14% and fat-
free
mass by 4.04 kg.
[76]
Giamatti
(Australia) 2014
Hypogonadal men
with type 2 diabetes
mellitus
(TT ≤ 300 ng/dL or
BioT ≤ 70 ng/dL)
88 TRT 45
placebo 43
TU 1000 mg/6–12
weeks intramuscular
for 56 weeks
TRT increased lean body mass. [77]
Stout
(UK) 2012
Men with chronic
heart failure
(TT < 15 nmol/L)
28 TRT 15
placebo 13
Testosterone 100
mg/2 weeks
intramuscular
for 12 weeks
(exercise)
TRT could not improve the shuttle
walk test and hand grip strength
compared with placebo.
[78]
Behre
(Australia) 2012
LOH men
(TT < 15 nmol/L or
BioT < 6.68 nmol/L
362 TRT 183
placebo 179
T gel 50–75 mg/day
for 6 months TRT increased lean body mass. [79]
Travison
(USA) 2011
Hypogonadal men
with mobility
limitation (TT 100–
350 ng/dL
or FT < 50 pg/mL)
209 TRT 106
placebo 103
T gel 100 mg/day
for 6 months
TRT increased leg-press and chest-
press strength and stair-climbing
power but could not improve walking
speed.
[80]
Atkinson
(UK) 2010 Hypogonadal frail
men (TT < 12 nmol/L)
30 TRT 16
placebo 14
T gel 50 mg/day
For 6 months.
TRT helped preserve muscle
thickness. There was no significant [81]
J. Clin. Med. 2022, 11, 6202 8 of 13
effect of treatment on fascicle length
or pennation angle.
Kenny
(USA) 2010
Hypogonadal frail
men
(TT < 350 ng/dL)
131 TRT 69
placebo 62
T gel 5 mg/day
for 12–24 months
There was an increase in lean mass in
the testosterone group but no
differences in strength or physical
performance.
[50]
Srinivas-
Shankar
(UK)
2010
Hypogonadal frail
men
(TT <
12 nmol/L or FT
< 250 pmol/L)
274 TRT 138
placebo 136
T gel 50 mg/day
for 6 months
Isometric knee extension peak torque
was improved, and lean mass was
increased in the TRT group.
[82]
TRT, testosterone replacement therapy; TT, total testosterone; FT, free testosterone; BioT, bioavaila-
ble testosterone; TU, testosterone undecanoate; TE, testosterone enanthate.
A recent meta-analysis demonstrated that TRT produced an increase in lean body
mass of 2.54 kg (95% CI 1.27–3.80; p < 0.001) and an increase in handgrip strength of 1.58
kg (95% CI 0.17–3.0; p = 0.03) and concluded that TRT showed a beneficial effect on sar-
copenic components, such as muscle mass and strength, as well as on physical perfor-
mance in middle-aged and older adults [83]. Another recent systematic review also sup-
ports certain beneficial contributions of TRT to muscle condition and function [84].
However, there are limited data currently available regarding the direct effects of
TRT on preventing sarcopenia, which is diagnosed based on muscle mass, strength, and
physical functions, and the conclusions drawn from those data have been conflicting
[50,69,72,80]. Two RCTs demonstrated that TRT contributed to an improvement in both
muscle mass/strength and physical performance [69,72]. However, a previous study that
included 209 hypogonadal men with mobility limitations reported that 6 months of TRT
could increase muscle strength and stair-climbing power but could not improve walking
speed [80]. Another study observed a significant increase in lean mass in the TRT group,
whereas there were no differences in strength or physical performance between the con-
trol and TRT groups [50]. Furthermore, clinical studies targeted to Asian people, espe-
cially men, have been extremely limited. Further studies with large numbers of partici-
pants and various races are likely to reach a more definite conclusion regarding the effects
of TRT on sarcopenia.
Studies have examined the dose-dependent effects of testosterone. A study that in-
cluded healthy male adult participants randomly assigned to a weekly administration
group (100 mg of testosterone enanthate weekly in an intramuscular injection) and a monthly
group (alternating months of 100 mg of testosterone or placebo) for 5 months demonstrated
that both groups had an increase in fiber diameter and peak power, with the weekly treatment
being five-fold more effective than the monthly treatment [85]. In addition, a higher dose of
testosterone can affect muscle mass and strength, not only for hypogonadal men but also eu-
gonadal older men and healthy young men [86]. These data suggest that the anabolic effects
of TRT are likely to be dose dependent to a certain extent. However, higher doses are associ-
ated with a high frequency of adverse effects and caution is required.
5.3. Other Systemic Effects of Testosterone Replacement Therapy
In general, TRT is rarely used to treat men solely for sarcopenia and is a widely ac-
cepted tool to improve various symptoms and clinical conditions occurred in hy-
pogonadal men, including decreased libido and sexual desire, depression, muscle weak-
ness, obesity, deterioration of insulin resistance, dyslipidemia, and osteoporosis [1–4].
Many RCTs and systematic reviews demonstrate that TRT can improve libido and sexual
function, mood and energy, quality of life, anemia, bone density, cognitive function, body
composition, in addition to muscle mass and strength [1,2,87–90]. In addition, some recent
studies have supported the long-term use of TRT for 4 to 5 years to obtain beneficial effects
for various metabolic parameters, body composition, and erectile function [91–94].
J. Clin. Med. 2022, 11, 6202 9 of 13
5.4. Adverse Effects and Risks of Testosterone Replacement Therapy
It is widely known that TRT is significantly associated with some adverse effects,
such as erythrocytosis, gynecomastia, liver toxicity, testicular atrophy and infertility, acne,
exacerbate sleep apnea, and potential growth of prostate cancer [1,87,88,95]. In particular,
elderly men who are often candidates for TRT are originally at increased risk of prostate
cancer. Therefore, men who receive TRT should undergo prostate-specific antigen screen-
ing regularly before and during treatment. On the other hand, there is no good evidence
that testosterone administration can convert subclinical prostate cancer to clinically sig-
nificant cancer and can increase risk of prostate cancer [87,95,96]. The association between
TRT and cardiovascular risk is still controversial. Some recent meta-analyses did not
demonstrate a significant association between TRT and any cardiovascular events [97,98].
6. Conclusions
Sarcopenia is an important pathophysiology factor of frailty in older adults and is
diagnosed in older adults with decreased muscle strength, muscle mass, and walking
speed, which can lead to a serious decrease in QOL. Testosterone directly interacts with
the androgen receptor expressed in myonuclei and satellite cells and is then also indirectly
associated with muscle metabolism through various cytokines and molecules. Significant
correlations between testosterone and frailty in men have been confirmed by numerous
cross-sectional studies. In addition, numerous RCTs have supported the beneficial effect
of TRT on muscle mass and strength among men with low to normal testosterone levels.
In the world’s aging society, TRT can be a tool for preventing the development of sarco-
penia in older-adult men, although further RCTs are required.
Author Contributions: Data collection, K.S. and Y.K.; writing—original draft preparation, K.S.;
writing—review and editing, K.S. and K.I.; supervision, A.M. All authors have read and agreed to
the published version of the manuscript.
Funding: This research received no external funding.
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement: Not applicable.
Conflicts of Interest: The authors declare no conflicts of interest.
References
1. Bassil, N.; Alkaade, S.; Morley, J.E. The benefits and risks of testosterone replacement therapy: A review. Ther. Clin. Risk Manag.
2009, 5, 427–448.
2. Harman, S.M.; Metter, E.J.; Tobin, J.D.; Pearson, J.; Blackman, M.R. Baltimore Longitudinal Study of Aging Baltimore longitu-
dinal study of aging: Longitudinal effects of aging on serum total and free testosterone levels in healthy men: Baltimore longi-
tudinal study of aging. J. Clin. Endocrinol. Metab. 2001, 86, 724–731.
3. Lunenfeld, B.; Mskhalaya, G.; Zitzmann, M.; Arver, S.; Kalinchenko, S.; Tishova, Y.; Morgentaler, A. Recommendations on the
diagnosis, treatment and monitoring of hypogonadism in men. Aging Male 2015, 18, 5–15.
4. Lunenfeld, B.; Arver, S.; Moncada, I.; Rees, D.A.; Schulte, H.M. How to help the aging male? Current approaches to hypogonad-
ism in primary care. Aging Male 2012, 15, 187–197.
5. Delmonico, M.J.; Harris, T.B.; Lee, J.S.; Visser, M.; Nevitt, M.; Kritchevsky, S.B.; Tylavsky, F.A.; Newman, A.B. Health, Aging
and Body Composition Study. Alternative definitions of sarcopenia, lower extremity performance, and functional impairment
with aging in older men and women. J. Am. Geriatr. Soc. 2007, 55, 769–774.
6. Xue, Q.L.; Bandeen-Roche, K.; Varadhan, R.; Zhou, J.; Fried, L.P. Initial manifestations of frailty criteria and the development
of frailty phenotype in the Women’s Health and Aging Study II. J. Gerontol. A Biol. Sci. Med. Sci. 2008, 63, 984–990.
7. Hida, T.; Harada, A.; Imagama, S.; Ishiguro, N. Managing sarcopenia and its related-fractures to improve quality of life in
geriatric populations. Aging Dis. 2014, 5, 226–237.
8. Kitamura, A.; Seino, S.; Abe, T.; Nofuji, Y.; Yokoyama, Y.; Amano, H.; Nishi, M.; Taniguchi, Y.; Narita, M.; Fujiwara, Y.; et al.
Sarcopenia: Prevalence, associated factors, and the risk of mortality and disability in Japanese older adults. J. Cachexia Sarcopenia
Muscle 2021, 12, 30–38.
J. Clin. Med. 2022, 11, 6202 10 of 13
9. Petermann-Rocha, F.; Balntzi, V.; Gray, S.R.; Lara, J.; Ho, F.K.; Pell, J.P.; Celis-Morales, C. Global prevalence of sarcopenia and
severe sarcopenia: A systematic review and meta-analysis. J. Cachexia Sarcopenia Muscle 2022, 13, 86–99.
10. Papadopoulou, S.K. Sarcopenia: A contemporary health problem among older adult populations. Nutrients 2020, 12, 1293.
11. Rosenberg, I.H. Sarcopenia: Origins and clinical relevance. J. Nutr. 1997, 127, 990S–991S.
12. Wannamethee, S.G.; Atkins, J.L. Muscle loss and obesity: The health implications of sarcopenia and sarcopenic obesity. Proc.
Nutr. Soc. 2015, 74, 405–412.
13. Baumgartner, R.N.; Koehler, K.M.; Gallagher, D.; Romero, L.; Heymsfield, S.B.; Ross, R.R.; Garry, P.J.; Lindeman, R.D. Epide-
miology of sarcopenia among the elderly in New Mexico. Am. J. Epidemiol. 1998, 147, 755–763.
14. Cruz-Jentoft, A.J.; Baeyens, J.P.; Bauer, J.M.; Boirie, Y.; Cederholm, T.; Landi, F.; Martin, F.C.; Michel, J.P.; Rolland, Y.; Schneider,
S.M.; et al. European Working Group on Sarcopenia in Older People Sarcopenia: European consensus on definition and diag-
nosis: Report of the European Working Group on Sarcopenia in Older People. Age Ageing 2010, 39, 412–423.
15. Studenski, S.A.; Peters, K.W.; Alley, D.E.; Cawthon, P.M.; McLean, R.R.; Harris, T.B.; Ferrucci, L.; Guralnik, J.M.; Fragala, M.S.;
Kenny, A.M.; et al. The FNIH sarcopenia project: Rationale, study description, conference recommendations, and final esti-
mates. J. Gerontol. A Biol. Sci. Med. Sci. 2014, 69, 547–558.
16. Chen, L.K.; Liu, L.K.; Woo, J.; Assantachai, P.; Auyeung, T.W.; Bahyah, K.S.; Chou, M.Y.; Chen, L.Y.; Hsu, P.S.; Krairit, O.; et al.
Sarcopenia in Asia: Consensus report of the Asian working group for sarcopenia. J. Am. Med. Dir. Assoc. 2014, 15, 95–101.
17. Yamada, M.; Nishiguchi, S.; Fukutani, N.; Tanigawa, T.; Yukutake, T.; Kayama, H.; Aoyama, T.; Arai, H. Prevalence of sarcope-
nia in community-dwelling Japanese older adults. J. Am. Med. Dir. Assoc. 2013, 14, 911–915.
18. Lin, C.C.; Lin, W.Y.; Meng, N.H.; Li, C.I.; Liu, C.S.; Lin, C.H.; Chang, C.K.; Lee, Y.D.; Lee, C.C.; Li, T.C. Sarcopenia prevalence
and associated factors in an elderly Taiwanese metropolitan population. J. Am. Geriatr. Soc. 2013, 61, 459–462.
19. Cruz-Jentoft, A.J.; Landi, F.; Schneider, S.M.; Zúñiga, C.; Arai, H.; Boirie, Y.; Chen, L.K.; Fielding, R.A.; Martin, F.C.; Michel, J.P.;
et al. Prevalence of and interventions for sarcopenia in ageing adults: A systematic review. Report of the international sarcope-
nia initiative (EWGSOP and IWGS). Age Ageing 2014, 43, 748–759.
20. Papadopoulou, S.K.; Tsintavis, P.; Potsaki, P.; Papandreou, D. Differences in the prevalence of sarcopenia in community-dwell-
ing, nursing home and hospitalized individuals. A systematic review and meta-analysis. J. Nutr. Health Aging 2020, 24, 83–90.
21. Vellas, B.; Guigoz, Y.; Garry, P.J.; Nourhashemi, F.; Bennahum, D.; Lauque, S.; Albarede, J.L. The Mini Nutritional Assessment
(MNA) and its use in grading the nutritional state of elderly patients. Nutrition 1999, 15, 116–122.
22. Walston, J.D. Sarcopenia in older adults. Curr. Opin. Rheumatol. 2012, 24, 623–627.
23. Buvat, J.; Maggi, M.; Guay, A.; Torres, L.O. Testosterone deficiency in men: Systematic review and standard operating proce-
dures for diagnosis and treatment. J. Sex. Med. 2013, 10, 245–284.
24. Kaufman, J.M.; Vermeulen, A. The decline of androgen levels in elderly men and its clinical and therapeutic implications. En-
docr. Rev. 2005, 26, 833–876.
25. Chamberlain, N.L.; Driver, E.D.; Miesfeld, R.L. The length and location of CAG trinucleotide repeats in the androgen receptor
N-terminal domain affect transactivation function. Nucleic Acids Res. 1994, 22, 3181–3186.
26. Lee, J.H.; Shin, J.H.; Park, K.P.; Kim, I.J.; Kim, C.M.; Lim, J.G.; Choi, Y.C.; Kim, D.S. Phenotypic variability in Kennedy’s disease:
Implication of the early diagnostic features. Acta. Neurol. Scand. 2005, 112, 57–63.
27. Dubois, V.; Laurent, M.; Boonen, S.; Vanderschueren, D.G.; Claessens, F. Androgens and skeletal muscle: Cellular and molecular
action mechanisms underlying the anabolic actions. Cell. Mol. Life Sci. 2012, 69, 1651–1667.
28. Powers, M.L.; Florini, J.R. A direct effect of testosterone on muscle cells in tissue culture. Endocrinology 1975, 97, 1043–1047.
29. Kadi, F. Cellular and molecular mechanisms responsible for the action of testosterone on human skeletal muscle. A basis for
illegal performance enhancement. Br. J. Pharmacol. 2008, 154, 522–528.
30. Demling, R.H.; Orgill, D.P. The anticatabolic and wound healing effects of the testosterone analog oxandrolone after severe
burn injury. J. Crit. Care 2000, 15, 12–17.
31. Mauras, N.; Hayes, V.; Welch, S.; Rini, A.; Helgeson, K.; Dokler, M.; Veldhuis, J.D.; Urban, R.J. Testosterone deficiency in young
men: Marked alterations in whole body protein kinetics, strength, and adiposity. J. Clin. Endocrinol. Metab. 1998, 83, 1886–1892.
32. Sinha-Hikim, I.; Cornford, M.; Gaytan, H.; Lee, M.L.; Bhasin, S. Effects of testosterone supplementation on skeletal muscle fiber
hypertrophy and satellite cells in community-dwelling older men. J. Clin. Endocrinol. Metab. 2006, 91, 3024–3033.
33. Sculthorpe, N.; Solomon, A.M.; Sinanan, A.C.; Bouloux, P.M.; Grace, F.; Lewis, M.P. Androgens affect myogenesis in vitro and
increase local IGF-1 expression. Med. Sci. Sports Exerc. 2012, 44, 610–615.
34. Bhasin, S.; Woodhouse, L.; Casaburi, R.; Singh, A.B.; Bhasin, D.; Berman, N.; Chen, X.; Yarasheski, K.E.; Magliano, L.; Dzekov, C.; et
al. Testosterone dose-response relationships in healthy young men. Am. J. Physiol. Endocrinol. Metab. 2001, 281, E1172–E1181.
35. Singh, R.; Bhasin, S.; Braga, M.; Artaza, J.N.; Pervin, S.; Taylor, W.E.; Krishnan, V.; Sinha, S.K.; Rajavashisth, T.B.; Jasuja, R.
Regulation of myogenic differentiation by androgens: Cross talk between androgen receptor/beta-catenin and follistatin/trans-
forming growth factor-beta signaling pathways. Endocrinology 2009, 150, 1259–1268.
36. Qin, W.; Pan, J.; Wu, Y.; Bauman, W.A.; Cardozo, C. Protection against dexamethasone-induced muscle atrophy is related to
modulation by testosterone of FOXO1 and PGC-1alpha. Biochem. Biophys. Res. Commun. 2010, 403, 473–478.
37. Wu, Y.; Bauman, W.A.; Blitzer, R.D.; Cardozo, C. Testosterone-induced hypertrophy of L6 myoblasts is dependent upon Erk
and mTOR. Biochem. Biophys. Res. Commun. 2010, 400, 679–683.
38. Pires-Oliveira, M.; Maragno, A.L.; Parreiras-e-Silva, L.T.; Chiavegatti, T.; Gomes, M.D.; Godinho, R.O. Testosterone represses ubiquitin
ligases atrogin-1 and Murf-1 expression in an androgen-sensitive rat skeletal muscle in vivo. J. Appl. Physiol. 2010, 108, 266–273.
J. Clin. Med. 2022, 11, 6202 11 of 13
39. Morley, J.E.; Baumgartner, R.N.; Roubenoff, R.; Mayer, J.; Nair, K.S. Sarcopenia. J. Lab. Clin. Med. 2001, 137, 231–243.
40. Smith, M.R. Changes in fat and lean body mass during androgen-deprivation therapy for prostate cancer. Urology 2004, 63, 742–745.
41. Chang, D.; Joseph, D.J.; Ebert, M.A.; Galvão, D.A.; Taaffe, D.R.; Denham, J.W.; Newton, R.U.; Spry, N.A. Effect of androgen
deprivation therapy on muscle attenuation in men with prostate cancer. J. Med. Imaging Radiat. Oncol. 2014, 58, 223–228.
42. Mitsuzuka, K.; Arai, Y. Metabolic changes in patients with prostate cancer during androgen deprivation therapy. Int. J. Urol.
2018, 25, 45–53.
43. Baumgartner, R.N.; Waters, D.L.; Gallagher, D.; Morley, J.E.; Garry, P.J. Predictors of skeletal muscle mass in elderly men and
women. Mech. Ageing Dev. 1999, 107, 123–136.
44. van den Beld, A.; Huhtaniemi, I.T.; Pettersson, K.S.; Pols, H.A.; Grobbee, D.E.; de Jong, F.H.; Lamberts, S.W. Luteinizing hormone and
different genetic variants, as indicators of frailty in healthy elderly men. J. Clin. Endocrinol. Metab. 1999, 84, 1334–1339.
45. Szulc, P.; Duboeuf, F.; Marchand, F.; Delmas, P.D. Hormonal and lifestyle determinants of appendicular skeletal muscle mass
in men: The MINOS study. Am. J. Clin. Nutr. 2004, 80, 496–503.
46. Mouser, J.G.; Loprinzi, P.D.; Loenneke, J.P. The association between physiologic testosterone levels, lean mass, and fat mass in
a nationally representative sample of men in the United States. Steroids 2016, 115, 62–66.
47. Kong, S.H.; Kim, J.H.; Lee, J.H.; Hong, A.R.; Shin, C.S.; Cho, N.H. Dehydroepiandrosterone sulfate and free testosterone but not
estradiol are related to muscle strength and bone microarchitecture in older adults. Calcif. Tissue Int. 2019, 105, 285–293.
48. Ye, J.; Zhai, X.; Yang, J.; Zhu, Z. Association between serum testosterone levels and body composition among men 20–59 years
of age. Int. J. Endocrinol. 2021, 2021, 7523996.
49. Laurent, M.R.; Dedeyne, L.; Dupont, J.; Mellaerts, B.; Dejaeger, M.; Gielen, E. Age-related bone loss and sarcopenia in men.
Maturitas 2019, 122, 51–56.
50. Kenny, A.M.; Kleppinger, A.; Annis, K.; Rathier, M.; Browner, B.; Judge, J.O.; McGee, D. Effects of transdermal testosterone on
bone and muscle in older men with low bioavailable testosterone levels, low bone mass, and physical frailty. J. Am. Geriatr. Soc.
2010, 58, 1134–1143.
51. Wittert, G.A.; Chapman, I.M.; Haren, M.T.; Mackintosh, S.; Coates, P.; Morley, J.E. Oral testosterone supplementation increases
muscle and decreases fat mass in healthy elderly males with low-normal gonadal status. J. Gerontol. A Biol. Sci. Med. Sci. 2003,
58, 618–625.
52. Mohr, B.A.; Bhasin, S.; Kupelian, V.; Araujo, A.B.; O’Donnell, A.B.; McKinlay, J.B. Testosterone, sex hormone binding globulin,
and frailty in older men. J. Am. Geriatr. Soc. 2007, 55, 548–555.
53. Carcaillon, L.; Blanco, C.; Alonso-Bouzón, C.; Alfaro-Acha, A.; Garcia-García, F.J.; Rodriguez-Mañas, L. Sex differences in the
association between serum levels of testosterone and frailty in an elderly population: The Toledo Study for Healthy Aging. PLoS
ONE 2012, 7, e32401.
54. Schaap, L.A.; Pluijm, S.M.F.; Smit, J.H.; van Schoor, N.M.; Visser, M.; Gooren, L.J.G.; Lips, P. The association of sex hormonelevels with
poor mobility, low muscle strength and incidence of falls amongolder men and women. Clin. Endocrinol. 2005, 63, 152–160.
55. Eichholzer, M.; Barbir, A.; Basaria, S.; Dobs, A.S.; Feinleib, M.; Guallar, E.; Menke, A.; Nelson, W.G.; Rifai, N.; Platz, E.A.; et al.
Serum sex steroid hormones and frailty in older American men of the Third National Health and Nutrition Examination Survey
(NHANES III). Aging Male 2012, 15, 208–215.
56. Peng, X.; Hou, L.; Zhao, Y.; Lin, T.; Wang, H.; Gao, L.; Yue, J. Frailty and testosterone level in older adults: A systematic review
and meta-analysis. Eur. Geriatr. Med. 2022, 13, 663–673.
57. Yuki, A.; Otsuka, R.; Kozakai, R.; Kitamura, I.; Okura, T.; Ando, F.; Shimokata, H. Relationship between low free testosterone
levels and loss of muscle mass. Sci. Rep. Sci. Rep. 2013, 3, 1818.
58. Krasnoff, J.B.; Basaria, S.; Pencina, M.J.; Jasuja, G.K.; Vasan, R.S.; Ulloor, J.; Zhang, A.; Coviello, A.; Kelly-Hayes, M.; D’Agostino,
R.B.; et al. Free testosterone levels are associated with mobility limitation and physical performance in community-dwelling
men: The Framingham Offspring Study. J. Clin. Endocrinol. Metab. 2010, 95, 2790–2799.
59. Schaap, L.A.; Pluijm, S.M.F.; Deeg, D.J.H.; Penninx, B.W.; Nicklas, B.J.; Lips, P.; Harris, T.B.; Newman, A.B.; Kritchevsky, S.B.;
Cauley, J.A.; et al. Health ABC study. Low testosterone levels and decline in physical performance and muscle strength in older
men: Findings from two prospective cohort studies. Clin. Endocrinol. 2008, 68, 42–50.
60. Shin, Y.S.; Park, J.K. The Optimal Indication for Testosterone Replacement Therapy in Late Onset Hypogonadism. J. Clin. Med.
2019, 8, 209.
61. Mulhall, J.P.; Trost, L.W.; Brannigan, R.E.; Kurtz, E.G.; Redmon, J.B.; Chiles, K.A.; Lightner, D.J.; Miner, M.M.; Murad, M.H.;
Nelson, C.J.; et al. Evaluation and Management of Testosterone Deficiency: AUA Guideline. J. Urol. 2018, 200, 423–432.
62. Nieschlag, E.; Swerdloff, R.; Behre, H.M.; Gooren, L.J.; Kaufman, J.M.; Legros, J.J.; Lunenfeld, B.; Morley, J.E.; Schulman, C.;
Wang, C.; et al. Investigation, treatment, and monitoring of late-onset hypogonadism in males: ISA, ISSAM, and EAU recom-
mendations. J. Androl. 2006, 27, 135–137.
63. Dean, J.D.; McMahon, C.G.; Guay, A.T.; Morgentaler, A.; Althof, S.E.; Becher, E.F.; Bivalacqua, T.J.; Burnett, A.L.; Buvat, J.; El
Meliegy, A.; et al. The International Society for Sexual Medicine’ s Process of Care for the Assessment and Management of
Testosterone Deficiency in Adult Men. J. Sex. Med. 2015, 12, 1660–1686.
64. Hackett, G.; Kirby, M.; Edwards, D.; Jones, T.H.; Wylie, K.; Ossei-Gerning, N.; David, J.; Muneer, A. British Society for Sexual
Medicine Guidelines on Adult Testosterone Deficiency, with Statements for UK Practice. J. Sex. Med. 2017, 14, 1504–1523.
65. Kolind, M.I.; Christensen, L.L.; Caserotti, P.; Andersen, M.S.; Glintborg, D. Muscle function following testosterone replacement
in men on opioid therapy for chronic non-cancer pain: A randomized controlled trial. Andrology 2022, 10, 551–559.
J. Clin. Med. 2022, 11, 6202 12 of 13
66. Barnouin, Y.; Armamento-Villareal, R.; Celli, A.; Jiang, B.; Paudyal, A.; Nambi, V.; Bryant, M.S.; Marcelli, M.; Garcia, J.M.;
Qualls, C.; et al. Testosterone replacement therapy added to intensive lifestyle intervention in older men with obesity and hy-
pogonadism. J. Clin. Endocrinol. Metab. 2021, 106, e1096–e1110.
67. Chasland, L.C.; Yeap, B.B.; Maiorana, A.J.; Chan, Y.X.; Maslen, B.A.; Cooke, B.R.; Dembo, L.; Naylor, L.H.; Green, D.J. Testos-
terone and exercise: Effects on fitness, body composition, and strength in middle-to-older aged men with low-normal serum
testosterone levels. Am. J. Physiol. Heart Circ. Physiol. 2021, 320, H1985–H1998.
68. Glintborg, D.; Vaegter, H.B.; Christensen, L.L.; Bendix, E.; Graven-Nielsen, T.; Andersen, P.G.; Andersen, M. Testosterone re-
placement therapy of opioid-induced male hypogonadism improved body composition but not pain perception: A double-
blind, randomized, and placebo-controlled trial. Eur. J. Endocrinol. 2020, 182, 539–548.
69. Gagliano-Jucá, T.; Storer, T.W.; Pencina, K.M.; Travison, T.G.; Li, Z.; Huang, G.; Hettwer, S.; Dahinden, P.; Bhasin, S.; Basaria,
S. Testosterone does not affect agrin cleavage in mobility-limited older men despite improvement in physical function. Androl-
ogy 2018, 6, 29–36.
70. Storer, T.W.; Basaria, S.; Traustadottir, T.; Harman, S.M.; Pencina, K.; Li, Z.; Travison, T.G.; Miciek, R.; Tsitouras, P.; Hally, K.;
et al. Effects of testosterone supplementation for 3 years on muscle performance and physical function in older men. J. Clin.
Endocrinol. Metab. 2017, 102, 583–593.
71. Ng Tang Fui, M.; Prendergast, L.A.; Dupuis, P.; Raval, M.; Strauss, B.J.; Zajac, J.D.; Grossmann, M. Effects of testosterone treat-
ment on body fat and lean mass in obese men on a hypocaloric diet: A randomised controlled trial. BMC Med. 2016, 14, 153.
72. Dias, J.P.; Melvin, D.; Simonsick, E.M.; Carlson, O.; Shardell, M.D.; Ferrucci, L.; Chia, C.W.; Basaria, S.; Egan, J.M. Effects of aromatase
inhibition vs. testosterone in older men with low testosterone: Randomized-controlled trial. Andrology 2016, 4, 33–40.
73. Konaka, H.; Sugimoto, K.; Orikasa, H.; Iwamoto, T.; Takamura, T.; Takeda, Y.; Shigehara, K.; Iijima, M.; Koh, E.; Namiki, M.;
EARTH Study Group. Effects of long-term androgen replacement therapy on the physical and mental statuses of aging males with
late-onset hypogonadism: A multicenter randomized controlled trial in Japan (EARTH Study). Asian J. Androl. 2016, 18, 25–34.
74. Magnussen, L.V.; Glintborg, D.; Hermann, P.; Hougaard, D.M.; Højlund, K.; Andersen, M. Effect of testosterone on insulin
sensitivity, oxidative metabolism and body composition in aging men with type 2 diabetes on metformin monotherapy. Diabetes
Obes. Metab. 2016, 18, 980–989.
75. Sinclair, M.; Grossmann, M.; Hoermann, R.; Angus, P.W.; Gow, P.J. Testosterone therapy increases muscle mass in men with
cirrhosis and low testosterone: A randomised controlled trial. J. Hepatol. 2016, 65, 906–913.
76. Borst, S.E.; Yarrow, J.F.; Conover, C.F.; Nseyo, U.; Meuleman, J.R.; Lipinska, J.A.; Braith, R.W.; Beck, D.T.; Martin, J.S.; Morrow,
M.; et al. Musculoskeletal and prostate effects of combined testosterone and finasteride administration in older hypogonadal
men: A randomized, controlled trial. Am. J. Physiol. Endocrinol. Metab. 2014, 306, E433–E442.
77. Gianatti, E.J.; Dupuis, P.; Hoermann, R.; Strauss, B.J.; Wentworth, J.M.; Zajac, J.D.; Grossmann, M. Effect of testosterone treat-
ment on glucose metabolism in men with type 2 diabetes: A randomized controlled trial. Diabetes Care 2014, 37, 2098–2107.
78. Stout, M.; Tew, G.A.; Doll, H.; Zwierska, I.; Woodroofe, N.; Channer, K.S.; Saxton, J.M. Testosterone therapy during exercise
rehabilitation in male patients with chronic heart failure who have low testosterone status: A double-blind randomized con-
trolled feasibility study. Am. Heart J. 2012, 164, 893–901.
79. Behre, H.M.; Tammela, T.L.; Arver, S.; Tolrá, J.R.; Bonifacio, V.; Lamche, M.; Kelly, J.; Hiemeyer, F.; European Testogel® Study
Team; Giltay, E.J.; et al. A randomized, double-blind, placebo-controlled trial of testosterone gel on body composition and
health-related quality-of-life in men with hypogonadal to low-normal levels of serum testosterone and symptoms of androgen
deficiency over 6 months with 12 months open-label follow-up. Aging Male 2012, 15, 198–207.
80. Travison, T.G.; Basaria, S.; Storer, T.W.; Jette, A.M.; Miciek, R.; Farwell, W.R.; Choong, K.; Lakshman, K.; Mazer, N.A.; Coviello,
A.D.; et al. Clinical meaningfulness of the changes in muscle performance and physical function associated with testosterone
administration in older men with mobility limitation. J. Gerontol. A Biol. Sci. Med. Sci. 2011, 66, 1090–1099.
81. Atkinson, R.A.; Srinivas-Shankar, U.; Roberts, S.A.; Connolly, M.J.; Adams, J.E.; Oldham, J.A.; Wu, F.C.; Seynnes, O.R.; Stewart,
C.E.; Maganaris, C.N.; et al. Effects of testosterone on skeletal muscle architecture in intermediate-frail and frail elderly men. J.
Gerontol. A Biol. Sci. Med. Sci. 2010, 65, 1215–1219.
82. Srinivas-Shankar, U.; Roberts, S.A.; Connolly, M.J.; O’Connell, M.D.; Adams, J.E.; Oldham, J.A.; Wu, F.C. Effects of testosterone
on muscle strength, physical function, body composition, and quality of life in intermediate-frail and frail elderly men: A ran-
domized, double-blind, placebo-controlled study. J. Clin. Endocrinol. Metab. 2010, 95, 639–650.
83. Parahiba, S.M.; Ribeiro, É.C.T.; Corrêa, C.; Bieger, P.; Perry, I.S.; Souza, G.C. Effect of testosterone supplementation on sarcope-
nic components in middle-aged and elderly men: A systematic review and meta-analysis. Exp. Gerontol. 2020, 142, 111106.
84. Shin, M.J.; Jeon, Y.K.; Kim, I.J. Testosterone and sarcopenia. World J. Mens Health 2018, 36, 192–198.
85. Fitts, R.H.; Peters, J.R.; Dillon, E.L.; Durham, W.J.; Sheffield-Moore, M.; Urban, R.J. Weekly versus monthly testosterone admin-
istration on fast and slow skeletal muscle fibers in older adult males. J. Clin. Endocrinol. Metab. 2015, 100, E223–E231.
86. Bhasin, S.; Woodhouse, L.; Casaburi, R.; Singh, A.B.; Mac, R.P.; Lee, M.; Yarasheski, K.E.; Sinha-Hikim, I.; Dzekov, C.; Dzekov,
J.; et al. Older men are as responsive as young men to the anabolic effects of graded doses of testosterone on the skeletal muscle.
J. Clin. Endocrinol. Metab. 2005, 90, 678–688.
87. Hisasue, S. Contemporary perspective and management of testosterone deficiency: Modifiable factors and variable manage-
ment. Int. J. Urol. 2015, 22, 1084–1095.
88. Barbonetti, A.; D’Andrea, S.; Francavilla, S. Testosterone replacement therapy. Andrology 2020, 8, 1551–1566.
J. Clin. Med. 2022, 11, 6202 13 of 13
89. Shigehara, K.; Izumi, K.; Kadono, Y.; Mizokami, A. Testosterone and Bone Health in Men: A Narrative Review. J. Clin. Med.
2021, 10, 530.
90. Bhasin, S. Testosterone replacement in aging men: An evidence-based patient-centric perspective. J. Clin. Investig. 2021, 131,
e146607.
91. Francomano, D.; Lenzi, A.; Aversa, A. Effects of five-year treatment with testosterone undecanoate on metabolic and hormonal
parameters in ageing men with metabolic syndrome. Int. J. Endocrinol. 2014, 2014, 527470.
92. Traish, A.M.; Haider, A.; Doros, G.; Saad, F. Long-term testosterone therapy in hypogonadal men ameliorates elements of the
metabolic syndrome: An observational, long-term registry study. Int. J. Clin. Pract. 2014, 68, 314–329.
93. Hackett, G.; Cole, N.; Mulay, A.; Strange, R.C.; Ramachandran, S. Long-term testosterone therapy in type 2 diabetes is associated
with decreasing waist circumference and improving erectile function. World J. Mens. Health. 2020, 38, 68–77.
94. Kato, Y.; Shigehara, K.; Nakashima, K.; Iijima, M.; Kawagushi, S.; Nohara, T.; Izumi, K.; Kadono, Y.; Konaka, H.; Namiki, M.; et
al. The five-year effects of testosterone replacement therapy on lipid profile and glucose tolerance among hypogonadal men in
Japan: A case control study. Aging Male 2020, 23, 23–28.
95. Rodrigues dos Santos, M.; Bhasin, S. Benefits and Risks of Testosterone Treatment in Men with Age-Related Decline in Testos-
terone. Annu. Rev. Med. 2021, 72, 75–91.
96. Shabsigh, R.; Crawford, E.D.; Nehra, A.; Slawin, K.M. Testosterone therapy in hypogonadal men and potential prostate cancer
risk: A systematic review. Int. J. Impot. Res. 2009, 21, 9–23.
97. Alexander, G.C.; Iyer, G.; Lucas, E.; Lin, D.; Singh, S. Cardiovascular Risks of Exogenous Testosterone Use Among Men: A
Systematic Review and Meta-Analysis. Am. J. Med. 2017, 130, 293–305.
98. Corona, G.; Rastrelli, G.; Di Pasquale, G.; Sforza, A.; Mannucci, E.; Maggi, M. Testosterone and Cardiovascular Risk: Meta-
Analysis of Interventional Studies. J. Sex. Med. 2018, 15, 820–838.
... Free testosterone (FT), the active form of testosterone taken up by target cells, binds to the androgen receptor (AR). Several ARs expressed in muscle play important roles in maintaining muscle strength and muscle mass [6,7]. Serum testosterone levels decrease by 2-3% annually with age in men [8]. ...
... Although a few studies have failed to show a positive effect of testosterone on muscle strength [25,26], the current evidence has established a negative effect of low testosterone levels on muscle mass and strength [6]. In one cross-sectional study, serum FT levels were significantly correlated with appendicular muscle mass [9]. ...
... Low FT levels also cause muscle atrophy via both decreased anabolic and increased catabolic effects. Decreased anabolic effects are mediated by reduced myogenesis, differentiation of satellite cells, and protein synthesis, and increased catabolic effects are associated with enhanced muscle atrogin-1 and RING-finger protein-1 expression [6]. In addition, visceral fat deposition due to low FT levels contributes to an increase in inflammatory cytokines, such as interleukin-6 and tumor necrosis factor, which have catabolic effects on skeletal muscles [36]. ...
Article
Full-text available
Background and Objectives: Sarcopenic obesity, a clinical condition coexisting with obesity and sarcopenia, is associated with a high risk of functional impairment, reduced quality of life, and increased mortality. A decline in age-related free testosterone (FT) levels has been reported to be associated with decreased muscle mass and muscle strength and increased fat mass. However, the association between low FT levels and risk of sarcopenic obesity has not been well studied. This study aimed to investigate the direct association between low FT levels and sarcopenic obesity. Materials and Methods: This cross-sectional study used data of 982 community-dwelling men aged 70–84 years from the Korean Frailty and Aging Cohort Study. Sarcopenia was defined according to the criteria of the Asian Group for Sarcopenia (AWGS) 2019. Obesity was defined as a body fat mass ≥28.3%. Participants who met both sarcopenia and obesity criteria were defined as having sarcopenic obesity. Low FT levels were defined as FT levels <17.35 pmol/L according to the Endocrine Society Clinical Practice Guidelines. Results: The prevalence of sarcopenia, obesity, and sarcopenic obesity was significantly higher in the low-FT group than in the normal-FT group. Low FT levels were significantly associated with a higher risk of obesity (odds ratio [OR], 2.09, 95% confidence interval [CI], 1.11–3.92), sarcopenia (2.57, 95% CI 1.08–6.10), and sarcopenic obesity (3.66, 95% CI 1.58–8.47) compared with the healthy control group. The risk of low appendicular skeletal muscle mass index (ASMI) (1.78, 95% CI 1.04–3.02) and high fat mass (1.92, 95% CI 1.12–3.31) was significantly higher in the low-FT group than in the normal-FT group. Conclusions: This study showed that low FT levels were associated with a higher risk of sarcopenic obesity. Low FT levels were mainly related to body composition parameters such as low ASMI and high fat mass.
... Similarly, studies claimed that men are faster in repeating a single movement, while women outperform men in the speed of programming a sequence of hand movements (Nicholson and Kimura, 1996;Schmidt et al., 2000). It has been suggested that these performance patterns may be associated with testosterone-mediated fast-twitch muscle fibers or morphological asymmetry patterns in the brain, which were mainly described for the cerebellum (Fan et al., 2010;Hubel et al., 2013;Shigehara et al., 2022), an issue that warrants further investigation. Interestingly, there was no significant sex effect regarding the JTT. ...
Article
Full-text available
Age-related motor impairments often cause caregiver dependency or even hospitalization. However, comprehensive investigations of the different motor abilities and the changes thereof across the adult lifespan remain sparse. We, therefore, extensively assessed essential basic and complex motor functions in 444 healthy adults covering a wide age range (range 21 to 88 years). Basic motor functions, here defined as simple isolated single or repetitive movements in one direction, were assessed by means of maximum grip strength (GS) and maximum finger-tapping frequency (FTF). Complex motor functions, comprising composite sequential movements involving both proximal and distal joints/muscle groups, were evaluated with the Action Research Arm Test (ARAT), the Jebsen-Taylor Hand Function Test (JTT), and the Purdue Pegboard Test. Men achieved higher scores than women concerning GS and FTF, whereas women stacked more pins per time than men during the Purdue Pegboard Test. There was no significant sex effect regarding JTT. We observed a significant but task-specific reduction of basic and complex motor performance scores across the adult lifespan. Linear regression analyses significantly predicted the participants’ ages based on motor performance scores (R² = 0.502). Of note, the ratio between the left- and right-hand performance remained stable across ages for all tests. Principal Component Analysis (PCA) revealed three motor components across all tests that represented dexterity, force, and speed. These components were consistently present in young (21–40 years), middle-aged (41–60 years), and older (61–88 years) adults, as well as in women and men. Based on the three motor components, K-means clustering analysis differentiated high- and low-performing participants across the adult life span. The rich motor data set of 444 healthy participants revealed age- and sex-dependent changes in essential basic and complex motor functions. Notably, the comprehensive assessment allowed for generating robust motor components across the adult lifespan. Our data may serve as a reference for future studies of healthy subjects and patients with motor deficits. Moreover, these findings emphasize the importance of comprehensively assessing different motor functions, including dexterity, force, and speed, to characterize human motor abilities and their age-related decline.
... Aging also appears to induce a shift in the redox status towards oxidation, which is accompanied by overwhelmed or dysfunctional antioxidant defense mechanisms, ensuing an amplified susceptibility of cellular and subcellular environments to damage [7]. Along with mitochondrial remodeling, an age-related decrease in the levels of sex steroid hormones in both women and men has been proposed as an important driver of sarcopenia [8,9]. In fact, these hormones can modulate via their receptors distinct signaling pathways in skeletal muscle, but the precise outcomes and sex disparities are not completely acknowledged, principally in skeletal muscle aging [10]. ...
Article
Sarcopenia is associated with reduced quality of life and premature mortality. The sex disparities in the processes underlying sarcopenia pathogenesis, which include mitochondrial dysfunction, are ill-understood and can be decisive for the optimization of sarcopenia-related interventions. To improve the knowledge regarding the sex differences in skeletal muscle aging, the gastrocnemius muscle of young and old female and male rats was analyzed with a focus on mitochondrial remodeling through the proteome profiling of mitochondria-enriched fractions. To the best of our knowledge, this is the first study analyzing sex differences in skeletal muscle mitochondrial proteome remodeling. Data demonstrated that age induced skeletal muscle atrophy and fibrosis in both sexes. In females, however, this adverse skeletal muscle remodeling was more accentuated than in males and might be attributed to an age-related reduction of 17beta-estradiol signaling through its estrogen receptor alpha located in mitochondria. The females-specific mitochondrial remodeling encompassed increased abundance of proteins involved in fatty acid oxidation, decreased abundance of the complexes subunits, and enhanced proneness to oxidative posttranslational modifications. This conceivable accretion of damaged mitochondria in old females might be ascribed to low levels of Parkin, a key mediator of mitophagy. Despite skeletal muscle atrophy and fibrosis, males maintained their testosterone levels throughout aging, as well as their androgen receptor content, and the age-induced mitochondrial remodeling was limited to increased abundance of pyruvate dehydrogenase E1 component subunit beta and electron transfer flavoprotein subunit beta. Herein, for the first time, it was demonstrated that age affects more severely the skeletal muscle mitochondrial proteome of females, reinforcing the necessity of sex-personalized approaches towards sarcopenia management, and the inevitability of the assessment of mitochondrion-related therapeutics. Open access | Full article here: https://doi.org/10.1016/j.freeradbiomed.2024.04.005
... In higher doses, it activates satellite cells and reduces adipose stem cells [158]. The dose-dependent anabolic effects were reported not only for hypogonadal men but also for eugonadal older men and healthy young men [166]. Alongside its almost ubiquitous, dose-dependent anabolic effect on muscle mass, the effects of testosterone replacement on muscle strength and function are also supported by recent studies. ...
Article
Sarcopenia is a skeletal muscle disorder characterized by progressive and generalized decline in muscle mass and function. Although it is mostly known as an age-related disorder, it can also occur secondary to systemic diseases such as malignancy or organ failure. It has demonstrated a significant relationship with adverse outcomes, e.g., falls, disabilities, and even mortality. Several breakthroughs have been made to find a pharmaceutical therapy for sarcopenia over the years, and some have come up with promising findings. Yet still no drug has been approved for its treatment. The key factor that makes finding an effective pharmacotherapy so challenging is the general paradigm of standalone/single diseases, traditionally adopted in medicine. Today, it is well known that sarcopenia is a complex disorder caused by multiple factors, e.g., imbalance in protein turnover, satellite cell and mitochondrial dysfunction, hormonal changes, low-grade inflammation, senescence, anorexia of aging, and behavioral factors such as low physical activity. Therefore, pharmaceuticals, either alone or combined, that exhibit multiple actions on these factors simultaneously will likely be the drug of choice to manage sarcopenia. Among various drug options explored throughout the years, testosterone still has the most cumulated evidence regarding its effects on muscle health and its safety. A mas receptor agonist, BIO101, stands out as a recent promising pharmaceutical. In addition to the conventional strategies (i.e., nutritional support and physical exercise), therapeutics with multiple targets of action or combination of multiple therapeutics with different targets/modes of action appear to promise greater benefit for the prevention and treatment of sarcopenia
Preprint
Full-text available
Purpose: We investigated the influence of sarcopenia on treatment outcomes in elderly patients receiving medical therapy for lower urinary tract symptoms (LUTS). Methods: We included male patients with LUTS aged ≥75 years who had not changed their medication for 1 year. Current conditions were estimated using IPSS, overactive bladder symptoms score (OABSS), and screening tool for sarcopenia (SARC-F). SARC-F score of ≧4 was defined as sarcopenia. Results: We included 59 patients in this study, including 16 patients (27.1%) with sarcopenia. IPSS total score, voiding symptom score of IPSS, and IPSS-QOL were significantly higher in the sarcopenia group than the non-sarcopenia group. The non-sarcopenia group had significantly larger PV (40 vs. 20 ml) and higher testosterone levels (497.5 vs. 369 ng/ml) compared with the sarcopenia group. Moderate and severe LUTS (IPSS total ≥8) persisted in 34 patients (57.6%) after medical treatment. Multivariate analysis showed that only SARC-F was a significant factor affecting the IPSS total ≥8 despite medical treatment for 1 year (OR 1.67, 95%CI: 1.13–2.46). Conclusion: Sarcopenia defined by using SARC-F is a significant factor for unsatisfactory medical treatment of LUTS in elderly men aged ≥75 years.
Article
Sarcopenia is a multifactorial condition characterized by loss of muscle mass. It poses significant health risks in older adults worldwide. Both pharmacological and non-pharmacological approaches are reported to address this disease. Certain dietary patterns, such as adequate energy intake and essential amino acids, have shown positive outcomes in preserving muscle function. Various medications, including myostatin inhibitors, growth hormones, and activin type II receptor inhibitors, have been evaluated for their effectiveness in managing sarcopenia. However, it is important to consider the variable efficacy and potential side effects associated with these treatments. There are currently no drugs approved by the Food and Drug Administration for sarcopenia. The ongoing research aims to develop more effective strategies in the future. Our review of research on disease mechanisms and drug development will be a valuable contribution to future research endeavors.
Article
Testosterone replacement therapy (TRT) is an indicated treatment of several medical conditions including late-onset hypogonadism, congenital syndromes, and gender affirmation hormonal therapy. Increasing population age, medical benefits, and public awareness of TRT have resulted in increased prevalence of its utilization. However, TRT is not without concern for adverse risks including venous thromboembolic complications, cardiovascular events, and prostate issues. In the field of orthopaedic surgery, research is beginning to delineate the complex relationship between TRT and the development of orthopaedic conditions and potential effects on surgical interventions and outcomes. In this review, we discuss current literature surrounding TRT and subsequent development of osteoarthritis, incidence of total joint arthroplasty, musculotendinous pathology, postoperative infection risk, improvements in postoperative rehabilitation metrics, enhancement of osseous healing, and increased bone-implant integration. The authors suggest future areas of investigation that may provide guidance on how surgeons can mitigate adverse risks while optimizing benefits of TRT in the orthopaedic patient.
Article
The efficacy of the NASA SPRINT exercise countermeasures program for quadriceps (vastus lateralis) and triceps surae (soleus) skeletal muscle health was investigated during 70 days of simulated microgravity. Individuals completed 6° head-down-tilt bedrest (BR, n=9), bedrest with resistance and aerobic exercise (BRE, n=9), or bedrest with resistance and aerobic exercise and low-dose testosterone (BRE+T, n=8). All groups were periodically tested for muscle (n=9 times) and aerobic (n=4 times) power during bedrest. In BR, surprisingly, the typical bedrest-induced decrements in vastus lateralis myofiber size and power were either blunted (MHC I) or eliminated (MHC IIa), along with no change (P>0.05) in %MHC distribution and blunted quadriceps atrophy. In BRE, MHC I (vastus lateralis and soleus) and IIa (vastus lateralis) contractile performance was maintained (P>0.05) or increased (P<0.05). Vastus lateralis hybrid fiber percentage was reduced (P<0.05) and energy metabolism enzymes and capillarization were generally maintained (P>0.05), while not all of these positive responses were observed in the soleus. Exercise offset 100% of quadriceps and ~ ⅔ of soleus whole muscle mass loss. Testosterone (BRE+T) did not provide any benefit over exercise alone for either muscle, and for some myocellular parameters appeared detrimental. In summary, the periodic testing likely provided a partial exercise countermeasure for the quadriceps in the bedrest group, which is a novel finding given the extremely low exercise dose. The SPRINT exercise program appears to be viable for the quadriceps; however, refinement is needed to completely protect triceps surae myocellular and whole muscle health for astronauts on long-duration spaceflights.
Article
Full-text available
Background Sarcopenia is defined as the loss of muscle mass and strength. Despite the seriousness of this disease, a single diagnostic criterion has not yet been established. Few studies have reported the prevalence of sarcopenia globally, and there is a high level of heterogeneity between studies, stemmed from the diagnostic criteria of sarcopenia and the target population. The aims of this systematic review and meta-analysis were (i) to identify and summarize the diagnostic criteria used to define sarcopenia and severe sarcopenia and (ii) to estimate the global and region-specific prevalence of sarcopenia and severe sarcopenia by sociodemographic factors. Methods Embase, MEDLINE, and Web of Science Core Collections were searched using relevant MeSH terms. The inclusion criteria were cross-sectional or cohort studies in individuals aged ≥18 years, published in English, and with muscle mass measured using dual-energy x-ray absorptiometry, bioelectrical impedance, or computed tomography (CT) scan. For the meta-analysis, studies were stratified by diagnostic criteria (classifications), cut-off points, and instruments to assess muscle mass. If at least three studies reported the same classification, cut-off points, and instrument to measure muscle mass, they were considered suitable for meta-analysis. Following this approach, 6 classifications and 23 subgroups were created. Overall pooled estimates with inverse-variance weights obtained from a random-effects model were estimated using the metaprop command in Stata. Results Out of 19 320 studies, 263 were eligible for the narrative synthesis and 151 for meta-analysis (total n = 692 056, mean age: 68.5 years). Using different classifications and cut-off points, the prevalence of sarcopenia varied between 10% and 27% in the studies included for meta-analysis. The highest and lowest prevalence were observed in Oceania and Europe using the European Working Group on Sarcopenia in Older People (EWGSOP) and EWGSOP2, respectively. The prevalence ranged from 8% to 36% in individuals <60 years and from 10% to 27% in ≥60 years. Men had a higher prevalence of sarcopenia using the EWGSOP2 (11% vs. 2%) while it was higher in women using the International Working Group on Sarcopenia (17% vs. 12%). Finally, the prevalence of severe sarcopenia ranged from 2% to 9%. Conclusions The prevalence of sarcopenia and severe sarcopenia varied considerably according to the classification and cut-off point used. Considering the lack of a single diagnostic for sarcopenia, future studies should adhere to current guidelines, which would facilitate the comparison of results between studies and populations across the globe.
Article
Full-text available
Introduction: Sex hormones play an important role in the development and maintenance of bone and muscle mass. However, studies regarding serum testosterone levels, osteoporosis, and sarcopenia in men are relatively sparse and have led to contradictory conclusions. Therefore, this study aimed to investigate the association between serum testosterone levels and body composition, including bone mineral density (BMD), appendicular lean mass index (ALMI), and appendicular fat mass index (AFMI), among men 20-59 years of age through a cross-sectional analysis of the National Health and Nutrition Examination Survey. Materials and methods: Our analysis was based on the data for 3,875 men, 20-59 years of age. Weighted multiple regression analyses were used to estimate the independent association between serum testosterone levels and body composition. Weighted generalized additive models and smooth curve fittings were used to characterize the nonlinear associations between them. Results: The association between the serum testosterone level and lumbar BMD was positive in each multivariable linear regression model. In the model adjusted for age and race, the serum testosterone level was negatively associated with ALMI. However, in the models adjusted for body mass index, this association became positive. In addition, the association between the serum testosterone level and AFMI was negative in each multivariable linear regression model. Conclusion: Our study demonstrated a positive association of serum testosterone level with lumbar BMD and ALMI, and a negative association with AFMI, among men 20-59 years of age, suggesting that increasing testosterone levels may be beneficial to skeletal health in young and middle-aged men with low testosterone levels.
Article
Full-text available
As men age, serum testosterone (T) concentrations decrease, as do fitness, strength and lean mass. Whether testosterone treatment confers additive benefit to reverse these changes when combined with exercise training in middle-to-older aged men remains unclear. We assessed the effects of T treatment and exercise, alone and in combination, on aerobic capacity (VO2peak), body composition and muscular strength in men 50-70yrs, waist circumference ≥95cm and low-normal serum T (6-14nmol·L-1). Participants (n=80) were randomised to AndroForte5® (Testosterone 5.0%w/v, 100mg/2mL) cream (T), or matching placebo (P), applied transdermally daily, and supervised centre-based exercise (Ex) or no additional exercise (NEx), for 12-weeks. Exercise increased VO2peak and strength vs non-exercise (VO2peak: T+Ex:+2.5, P+Ex:+3.2mL·kg-1·min-1, P<0.001; leg press: T+Ex:+31, P+Ex:+24kg, P=0.006). T treatment did not affect VO2peak or strength. Exercise decreased total (T+Ex:-1.7, P+Ex-2.3kg, P<0.001) and visceral fat (T+Ex:-0.1, P+Ex:-0.3kg, P=0.003), and increased total (T+Ex:+1.4, P+Ex:+0.7kg, P=0.008) and arm lean mass (T+Ex:+0.5, P+Ex:+0.3kg, P=0.024). T treatment did not affect total or visceral fat, but increased total (T+Ex:+1.4, T+NEx:+0.7kg, P=0.015), leg (T+Ex:+0.3, T+NEx:+0.2kg, P=0.024) and arm lean mass (T+Ex:+0.5, T+NEx:+0.2kg, P=0.046). T+Ex increased arm lean mass (T+Ex:+0.5kg vs P+NEx:-0.0kg, P=0.001) and leg strength (T+Ex:+31 vs P+NEx:+12kg, P=0.032) compared to P+NEx, with no other additive effects. Exercise training was more effective than T treatment in increasing aerobic capacity and decreasing total and visceral fat mass. T treatment at therapeutic doses increased lean mass but conferred limited additional benefit when combined with exercise. Exercise should be evaluated as an anti-ageing intervention in preference to testosterone treatment in men.
Article
Full-text available
Bone fracture due to osteoporosis is an important issue in decreasing the quality of life for elderly men in the current aging society. Thus, osteoporosis and bone fracture prevention is a clinical concern for many clinicians. Moreover, testosterone has an important role in maintaining bone mineral density (BMD) among men. Some testosterone molecular mechanisms on bone metabolism have been currently established by many experimental data. Concurrent with a decrease in testosterone with age, various clinical symptoms and signs associated with testosterone decline, including decreased BMD, are known to occur in elderly men. However, the relationship between testosterone levels and osteoporosis development has been conflicting in human epidemiological studies. Thus, testosterone replacement therapy (TRT) is a useful tool for managing clinical symptoms caused by hypogonadism. Many recent studies support the benefit of TRT on BMD, especially in hypogonadal men with osteopenia and osteoporosis, although a few studies failed to demonstrate its effects. However, no evidence supporting the hypothesis that TRT can prevent the incidence of bone fracture exists. Currently, TRT should be considered as one of the treatment options to improve hypogonadal symptoms and BMD simultaneously in symptomatic hypogonadal men with osteopenia.
Article
Full-text available
Background There is limited evidence on sarcopenia in Asian populations. This study aimed to clarify the prevalence, associated factors, and the magnitude of association with mortality and incident disability for sarcopenia and combinations of its components among Japanese community‐dwelling older adults. Methods We conducted a 5.8 year prospective study of 1851 Japanese residents aged 65 years or older (50.5% women; mean age 72.0 ± 5.9) who participated in health check‐ups. Sarcopenia was defined according to the Asian Working Group for Sarcopenia 2019 algorithm. Appendicular lean mass index (ALMI) was measured using direct segmental multi‐frequency bioelectrical impedance analysis. A Cox proportional hazards regression model was used to identify associations of sarcopenia and the combinations of its components with all‐cause mortality and incident disability. Results The prevalence of sarcopenia was 11.5% (105/917) in men and 16.7% (156/934) in women. Significant sarcopenia‐related factors other than ageing were hypoalbuminaemia, cognitive impairment, low activity, and recent hospitalization (all P‐values <0.05) among men and cognitive impairment (P = 0.004) and depressed mood (P < 0.001) among women. Individuals with sarcopenia had higher risks of mortality [hazard ratios (95% confidence interval): 2.0 (1.2–3.5) in men and 2.3 (1.1–4.9) in women] and incident disability [1.6 (1.0–2.7) in men and 1.7 (1.1–2.7) in women]. Compared with the individuals without any sarcopenia components, those having low grip strength and/or slow gait speed without low ALMI tended to have an increased risk of disability [1.4 (1.0–2.0), P = 0.087], but not mortality [1.3 (0.8–2.2)]. We did not find increased risks of these outcomes in participants having low ALMI in the absence of low grip strength and slow gait speed [1.2 (0.8–1.9) for mortality and 0.9 (0.6–1.3) for incident disability]. Conclusions Japanese older men and women meeting Asian criteria of sarcopenia had increased risks of all‐cause mortality and disability. There were no significant increased risks of death or incident disability for both participants with muscle weakness and/or low performance without low muscle mass and those with low muscle mass with neither muscle weakness nor low performance. Further studies are needed to examine the interaction between muscle loss, muscle weakness, and low performance for adverse health‐related outcomes.
Article
Purpose: We conducted a systematic review to evaluate the relationship between total testosterone (TT), free testosterone (fT), or sex hormone-binding globulin (SHBG) and frailty in older adults. Methods: We systematically searched nine databases (e.g. MEDLINE, Embase, ACP Journal Club, and the Cochrane library et al.) for papers on frailty and androgen levels published up to October 10, 2021. We calculated the odds ratio (OR) for the relationship between testosterone level and frailty by performing meta-analysis. Results: The search strategy yielded 311 hits in all databases combined. Eleven (seven cross-sectional studies and four cohort studies) met the inclusion criteria for meta-analysis. Among cross-sectional studies, meta-analysis revealed a significant association between TT and frailty in men (OR = 1.37 [95% CI 1.09, 1.72]) not women (OR = 1.06 [0.84, 1.34]). The fT was also significantly association with frailty in men (OR = 1.55 [1.06, 2.25] not women (OR = 1.35 [0.91, 2.01]). Cohort studies showed the same result in TT (OR = 1.09 [1.02, 1.18]) and fT (OR = 1.15 [1.02, 1.30]) for men. We did not find a significant association between SHBG and frailty. Conclusion: The findings of this systematic review and meta-analysis suggest that TT and fT were significantly associated with frailty in older men but not women.
Article
Background : Chronic pain and opioid treatment are associated with increased risk of male hypogonadism and subsequently decreased muscle function. A diagnosis of hypogonadism is based on the presence of low total testosterone (TT) and associated symptoms. The effect of testosterone replacement therapy (TRT) on muscle function in men with chronic pain and low TT remains to be investigated. Objectives : To investigate effects of TRT on muscle function and gait performance in men treated with opioids for chronic non-cancer pain. Materials and methods : Double-blind, placebo-controlled study. 41 men (>18 years) with opioid-treated chronic pain and serum total testosterone <12 nmol/L were randomized to 24 weeks TRT (Testosterone undecanoate injection three times/6 months, n = 20) or placebo injections (n = 21). Muscle function was measured as leg press maximal voluntary contraction (LP-MVC), leg extension power using the Nottingham power rig and handgrip strength using a handheld dynameter. Gait performance was measured at usual and maximal gait speed on a 10-m track. Body composition (lean body mass and fat mass) was determined by Dual-energy X-ray Absorptiometry. Mann-Whitney tests were performed on ∆-values (24–0 weeks) between TRT and placebo. Results : At baseline, median (interquartile range) age was 55 ± 13 years and BMI was 30.7 ± 5.2 kg/m². ∆-muscle function and ∆-gait performance were similar between TRT and placebo. Median ∆-LP-MVC was 174.2 ± 406.7 Newton following TRT and 7.6 ± 419.1 Newton after placebo, p = 0.091. ∆-lean body mass was significantly higher following TRT compared to placebo, 3.6 ± 2.7 vs 0.1 ± 3.5 kg, respectively (p <0.001). Discussion : TRT, compared to placebo, did not improve muscle function or gait performance despite increased lean body mass. Changes in body composition did not infer any changes in muscle function. Conclusion : 24 weeks TRT in opioid treated men with pain-related male hypogonadism did not improve muscle function. This article is protected by copyright. All rights reserved
Article
Background Obesity and hypogonadism additively contribute to frailty in older men; however, appropriate treatment remains controversial. Objective Determine whether testosterone replacement augments the effect of lifestyle therapy on physical function in older men with obesity and hypogonadism. Design Randomized, double-blind, placebo-controlled trial. Setting VA Medical Center Participants Eight-three older (age≥65 years) men with obesity (BMI≥30 kg/m 2) and persistently low AM testosterone (<10.4 nmol/L) associated with frailty. Interventions Participants were randomized to lifestyle therapy (weight management and exercise training) plus either testosterone (LT+Test) or placebo (LT+Pbo) for six months. Outcome Measures Primary outcome was change in Physical Performance Test (PPT) score. Secondary outcomes included other frailty measures, body composition, BMD, physical functions, hematocrit, PSA, and sex hormones. Results PPT score increased similarly in LT+Test and LT+Pbo group (17% vs. 16%; P=.58). VO2peak increased more in LT+Test than LT+Pbo (23% vs. 16%; P=.03). Despite similar -9% weight loss, lean body mass and thigh muscle volume decreased less in LT+Test than LT+Pbo (-2% vs. -3 %; P=.01 and -2% vs -4%; P=04). Hip BMD was preserved in LT+Test compared with LT+Pbo (0.5% vs. -1.1%; P=.003). Strength increased similarly in LT+Test and LT+Pbo (23% vs. 22%; P=.94). Hematocrit but not PSA increased more in LT+Test than LT+Pbo (5% vs 1%; P<.001). Testosterone levels increased more in LT+Test than LT+Pbo (167% vs 27%; P<.001). Conclusion In older, obese hypogonadal men, adding testosterone for six months to lifestyle therapy does not further improve overall physical function. However, our findings suggest that testosterone may attenuate the weight loss–induced reduction in muscle mass and hip BMD and may further improve aerobic capacity.
Article
The aim of this study was to conduct a systematic review of the literature of randomized controlled trials on the effect of testosterone (T) supplementation compared to the placebo group or lower dose on sarcopenic components (muscle mass, strength and physical performance) in middle-aged and elderly men. Major electronic databases were searched for articles published on or before December 2019. Studies including individuals with age ≥ 40 years and which described the effect of T supplementation on sarcopenic components were found eligible (11 studies). Outcomes were calculated as the difference in means between the experimental and control/placebo groups, and data were presented as effect size with 95% confidence limits (95%CI). The meta-analysis was performed using a random effects model. Regarding lean body mass (LBM), eight studies evaluated the effect of T supplementation on this outcome, of these, seven reported gains after the intervention period. Our meta-analysis showed a beneficial effect on LBM of 2.54 kg (95% CI, 1.27 to 3.80) (p < 0.001). In muscle strength (MS), seven included studies evaluated the handgrip strength (HGS) and just one reported gain after the intervention period, but the meta-analysis showed an increase for HGS of 1.58 kgf (95%CI, 0.17 to 3.0) (p = 0.03). The second outcome for MS was leg strength (LS), where nine studies were included and five demonstrated gains in this parameter after the intervention period. In the meta-analysis, two out of three tests showed an effect on LS: T supplementation increase the leg press strength in 91.23 N (95%CI, 0.23 to 182.22) (p = 0.05) and leg extension in 144.10 N (95%CI, 44.21 to 244.00) (p < 0.01). In physical performance, four studies evaluated this outcome, with three of them showing positive effects in this parameter. In the meta-analysis, only two studies that reported the same assessment test (Physical Performance Test) were included, but no effect of T supplementation on this parameter was found. It can be concluded that T supplementation influences sarcopenic components in middle-aged and older men, because is associated with increased in muscle mass and strength in addition to physical performance.